Next Article in Journal
Polarization of Melatonin-Modulated Colostrum Macrophages in the Presence of Breast Tumor Cell Lines
Previous Article in Journal
Adipose Tissue-Derived Products May Present Inflammatory Properties That Affect Chondrocytes and Synoviocytes from Patients with Knee Osteoarthritis
Previous Article in Special Issue
ATO Increases ROS Production and Apoptosis of Cells by Enhancing Calpain-Mediated Degradation of the Cancer Survival Protein TG2
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Transglutaminase in Foods and Biotechnology

1
Laboratory for Separation Processes and Product Design, Faculty of Chemistry and Chemical Engineering, University of Maribor, Smetanova Ulica 17, SI-2000 Maribor, Slovenia
2
Faculty of Medicine, University of Maribor, Taborska Ulica 8, SI-2000 Maribor, Slovenia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(15), 12402; https://doi.org/10.3390/ijms241512402
Submission received: 26 July 2023 / Revised: 1 August 2023 / Accepted: 2 August 2023 / Published: 3 August 2023

Abstract

:
Stabilization and reusability of enzyme transglutaminase (TGM) are important goals for the enzymatic process since immobilizing TGM plays an important role in different technologies and industries. TGM can be used in many applications. In the food industry, it plays a role as a protein-modifying enzyme, while, in biotechnology and pharmaceutical applications, it is used in mediated bioconjugation due to its extraordinary crosslinking ability. TGMs (EC 2.3.2.13) are enzymes that catalyze the formation of a covalent bond between a free amino group of protein-bound or peptide-bound lysine, which acts as an acyl acceptor, and the γ-carboxamide group of protein-bound or peptide-bound glutamine, which acts as an acyl donor. This results in the modification of proteins through either intramolecular or intermolecular crosslinking, which improves the use of the respective proteins significantly.

1. Introduction

Enzymes were used long before the development of modern DNA technology as fermenting microorganisms or crude preparations of different fruits. However, with the development of advanced bioprocesses using recombinant DNA technology, enzymes are being purified and produced on a larger scale, which has allowed their use and their applications in different industrial technologies, such as in the chemical, food, textile, cosmetic, pulp, and paper industries [1,2,3,4]. They are nutrients that play an important role in the physical properties of different foods. As such, they have been used widely in many industrial processes for the production of different products. Concerns regarding global food shortages and population growth, as well as the use of advanced food proteins, are increasing constantly in recent years. Moreover, the importance of protein modification technology is gaining much interest in order to meet the ever-challenging needs of a growing population [1,5,6,7].
The development of protein engineering with site-directed evolution has enabled novel enzymes with enhanced activities for many new processes, which makes industrial enzymes that are needed in everyday life more accessible to various industries [8,9]. Among the most-used industrial enzymes are hydrolases and carbohydrases. Hydrolases, such as lipases and proteases, are the dominant type used in many food industries, mostly dairy, as well as in the detergent and chemical industries. Carbohydrases include amylases and cellulases and are also used extensively. Table 1 lists some industrial enzymes with their significant industrial applications.
Drastic savings in resources were achieved by applying industrial enzymes in various processes; for example, energy efficiency and water and raw material consumption have improved significantly. Among other approaches, when compared to chemical modifications, applying enzymes in protein modification displays many advantages, which include high reaction specificities and low side-reaction frequencies, with the lack of need for high-pressure and high-temperature conditions. Such advantages make the protein modification technology effective, mostly in the food industry [10,11,12,13,14]. In terms of applying hydrolases in industry, proteases were the main protein-modifying enzymes. With the emergence of the enzyme TGM, which is involved in protein crosslinking, the protein modification technology possibilities have expanded enormously [15,16,17,18]. TGM (EC 2.3.2.13) belongs to the transferase family, distributed widely in nature. TGM is responsible for acyl transfer, deamidation, and crosslinking of intra- or inter-chain glutamine peptide moiety, which is the acyl donor and lysine peptide moiety, which is the acyl acceptor. The enzyme TGM catalyzes the addition of free amines into proteins by joining the glutamine residue. When the amine is absent, water becomes the acyl acceptor, and the γ-carboxamide groups deamidate to glutamic acid residues. The transamidation reaction occurs when the ε-amino groups of lysine residues in proteins act as acyl acceptors. In such cases, the acyl transfer onto a lysine residue forms intra-molecular and inter-molecular covalent crosslinks of ε-(γ-glutamyl)lysine, which is enriched with essential amino acids [19,20].
TGMs can be found in plants, such as soy, topinambour, and fodder beet; in animals, such as animal body fluids and fish; as well as in microorganisms. TGMs from mammalian sources are Ca2+-dependent, while microbial TGMs are Ca2+-independent and have smaller MW. Due to their Ca2+ independency, microbial TGMs are considered to be more cost-effective and eco-friendly, while their characteristics can prevent changes in formation of by-products, which occurs in Ca2+ protein complexes [21,22]. Moreover, their source origin also dictates their activities, which varies depending on their origin. The main differences between microbial TGM and existing TGMs from animal sources are presented in Table 2.
TGM-catalyzed reactions result in functional property changes, such as solubility foaming, viscosity, elasticity, water holding capacity, emulsifying capacity, gelation, and thermal stability of different food proteins [23,24,25]. TGM was also found to be involved in many physiological processes, such as in coagulation antibacterial immune reactions and in photosynthesis. TGM is an extracellular enzyme and was isolated from Streptoverticillium sp. And Physarum polycephalum. It can also be biosynthesized by many microorganisms, such as Streptoverticillium sp., Streptoverticillium cinnamoneum, Streptomyces netropsis, Streptoverticillium ladakanum, Streptomyces lydicus, and Bacillus subtilis.

2. Enzymatic Properties of TGMs

TGM modifies proteins with amine incorporation and crosslinking, where TGM catalyzes the reaction of the acyl transfer between the γ-carboxyamide group of peptides, which is bound with glutamine residue acyl donors and primary amines receptors of different compounds. The reaction can be found in Figure 1a. As presented in Figure 1b, the ε-amino group of lysine reacts as a receptor, which forms intra-molecular and inter-molecular crosslinks of ε-(γ-glutamyl)lysine isopeptides. When the lysine residue is absent, or when the protein system is free, water reacts as the receptor for the acyl groups, and the carboxyamide groups of the glutamine residues are deamidated, which, consequently, forms glutamic acid and ammonia residues that can modify protein charges and protein stability. The reaction is presented in Figure 1c.
The TGM crosslinking reaction occurs before the acyl transfer and deamidation reactions in food systems, which results in the formation of glutamyl lysine isopeptides and polymers with high molecular weight. Consequently, it changes the functional properties of the proteins, resulting in improved rheology and other quality properties for various food products (Figure 2) [26]. The properties and structure of TGM were studied by many researchers, where it was reported that the MW of microbial TGM is approximately 38,000 kDa and is only half of its MW when originating from an animal source [27,28]. The microbial TGM consists of 331 amino acids in only one polypeptide chain. The secondary structure of TGM consists of eight β-strands, which are surrounded by eleven α-helixes. Different 3D structures of TGM from different sources are presented in Figure 3. Microbial TGM has a stabile catalytic activity over a wider range of pH values in comparison to animal TGM. The optimal pH activity of microbial TGM is in the range 5–7 and has the isoelectric point of 8, while the optimal catalytic activity of TGM lies in the temperature range from 40–50 °C, at pH 6. When the temperature increases over 70 °C, the activity decreases drastically, resulting in activity loss [26,29,30].
The protein crosslinking reaction, called polymerization, can result in dimer, trimer, and polymer formation. For identifying these crosslinked formations, commonly used techniques are gel electrophoresis (GE), size exclusion chromatography (SEC), and isopeptide content quantification (ICQ). GE allows identification of casein molecules’ crosslinks, such as aS1-, aS2-, and κ-casein, whereas high-MW polymers with over 250 kDa cannot go through the gel [31]. The quantification of monomer conversions to dimer, trimer, and oligomer is identified by SEC, which provides an estimation of the polymerization degree (PD), also considered the crosslinking degree. The PD is the ratio between the dimer, trimer, and oligomer sum and the monomer, dimer, trimer, and oligomer sum. The ICQ technique can also quantify the crosslinking reaction since the isopeptide bonds form during the crosslinking reaction and do not succumb to protein hydrolysis [32,33].
Figure 3. TGMs and their 3D structures from animal source (Pagros major), microbial source (Streptomyces mobaraensis), and plant source (Phytophthora sojae) (RCSB Protein Data Bank [34]).
Figure 3. TGMs and their 3D structures from animal source (Pagros major), microbial source (Streptomyces mobaraensis), and plant source (Phytophthora sojae) (RCSB Protein Data Bank [34]).
Ijms 24 12402 g003

3. Origins of TGMs

Based on the similarities of catalytic properties and mechanism of TGM reactions, TGMs are believed to be evolutionary, close to thiol-like proteases. Clusters of TGM-like domains can be identified in eucaryotic TGMs that are found in all archaea and some yeast and bacterial species [35,36,37]. TGMs are enzymes found both on the inside and outside of the cell, which determines the versatility of their functions. Enzymatic activity of TGM was found in microorganisms, plant, and animal tissues. They all exhibit similar catalytic activity with biochemical properties, although those from plant and animal sources have less homology in the composition of amino acids [38,39,40]. TGMs from animal sources take part in many physiological processes, such as participating in skin formation, blood coagulation, and in antimicrobial immune reactions. Additionally, TGMs from plant sources play a role in the process of growth and development [41,42]. A specific feature of TGMs from plant sources is their sensitivity to light. This property applies especially to chloroplast TGM, which has been investigated by many studies [43,44]. Isolation and purification of TGM from microbiological sources has allowed its application in many processes and its simplification, which has provided economical savings with lower energy consumption. Gene transfer technology provided many possible TGM productions, where the expression of genes in E. coli has increased the production of TGMs and their efficiency immensely. Such enzymes are consumer-friendly and biodegradable, which offers a great advantage over many other chemical substances [45,46,47]. TGM is well-known for intra-molecular and inter-molecular formations of covalent bonds of glutamine and lysine, which initiate high-MW peptides, such as monomers, dimers, trimers, and oligomers. The digestibility of such crosslinked peptides has raised nutritional concerns. After ingesting crosslinked peptides, the dipeptide (glutamine–lysine) is cleaved by the activity of two enzymes. γ-glutamylamine cyclotransferase is a kidney enzyme cleaving the glutamine–lysine isopeptide, which yields free lysine and 5-oxo-proline, and is later metabolized to glutamate by 5-oxo-prolinase. γ-glutamine transpeptidase can be found in the intestinal membrane, kidneys, and blood. Microbial TGM is cultivated from Streptoverticillium strains [48,49,50,51]. However, the large amount of highly expensive nutrients makes its production not attractive from the economical angle. Many researchers have studied the use of agricultural waste materials as a source of carbon for TGM production. The media used to produce microbial TGM contain yeast extract, peptone, potassium and sodium phosphate, magnesium sulfate, and a source of carbon. The source of carbon may be xylose, which is a hemicellulose sugar used for bacterial proliferation [52]. The biosynthesis of TGM can be performed on different batch cultures, where the medium contains saccharose, glucose, starch, or dextrins as a carbon source. The culture Streptovitricillium mobaraense was found to be the most suitable medium, which contained corn steep, aminobac, and yeast extract as a source of nitrogen [53,54,55,56]. However, peptone, yeast extract, urea, and casein are used commonly as sources of nitrogen that are used in TGM biosynthesis. However, nowadays, TGM is usually produced by Streptovitricillium mobaraense in fermentation systems, which are followed by the downstream process presented in Figure 4. The main advantages of TGM production from microorganisms, compared to production from animal and plant sources, are the purity and high productivity since there is no need for difficult separation and filtration processes.
As eucaryotic TGMs are found in phylogenetic taxonomic groups, such as plants, animals, and fungi, the procaryotic TGMs are found in microorganisms. TGM was first isolated from S. mobaraensis in 1989, and later found in many other strains, listed in Table 3.
Microbial TGM from S. mobaraensis is used widely in different food industries, where it is applied in the reconstruction and manufacturing of meat, texturization of dairy products (yoghurt [42,89,90,91,92] and cheese [42,93,94,95,96]), or in different materials sciences, where it is used for the stabilization of wool and leather [97,98]. Due to the various applications of TGM, different grades of enzyme are available commercially. For biotechnological technologies, highly pure enzyme TGM is recommended to surpass by-products that can appear during side-reactions.

4. Applications of TGM

Enzyme TGM gained much attention due mainly to its potential for industrial applications. In their earlier uses, TGMs focused mostly on the cheesemaking processes, mainly improving the characteristics of the cheese itself. Later on, beverage properties were being improved by decreasing viscosity and solubility increases, as well as reduction in yoghurt syneresis. Therefore, the majority of investigations of applying TGMs in different processes focused mostly on improving the functional properties of proteins that can be utilized to develop improved food ingredients, such as crosslinked milk powders and high-added food products [33]. Such improvements were also possible due to the different immobilization techniques of TGM.

4.1. TGM Immobilization

The immobilization of TGM on solid supports is a widely used method to increase TGM stability and improve its spectrum of use and reuse. Site-specific modification is needed for such applications, especially when unstable targets are in question. Harsh conditions are usually applied during chemical immobilization. If compared to enzymatic catalysis, selective and fast performance in aqueous media is performed under mild process conditions, which has many advantages. Immobilizing the enzyme onto a given matrix is benefiting the enzyme with a stabile structure, which provides an advantage in resisting temperature and pH alterations by retaining catalytic activities [99,100,101,102]. Immobilized enzymes are more stable and have improved properties and features in terms of their kinetic aspects when compared to the free form of the enzyme. The enzymes’ improved features are due to the conformational changes that happen in the enzyme structure as a result of the chosen and most suitable immobilization method. In this manner, enhanced activity, stability, and selectivity can be observed [103,104,105,106]. For larger-scale applications, immobilized enzymes are the considered subject of choice. Not many studies were reported on immobilizing TGM. However, immobilized TGM was investigated on a few supports, such as thermo-responsive carboxylated poly (N-isopolylacrylamide), agarose beads, polypropylene microporous membranes, and various nanomaterials, such as magnetic nanoparticles (MNPs) or carbon nanotubes (CNTs), as well as in the form of crosslinked enzymes aggregates (CLEAs). For example, TGM was immobilized on multi-walled CNTs for tissue scaffold designing. The highest immobilization efficiency of 58% was achieved and a 4.8 fold increase in catalytic efficiency was observed [107]. Gianetto et al. reported on a new amperometric immunosensor, which was based on the covalent immobilization of TGM onto functionalized gold nanoparticles, and used for the determination of anti-tissue TGM antibodies in human serum [108]. Another piezoelectric immunosensor was developed for the detection of anti-tissue TGM antibodies as specific biomarkers for early diagnosis of celiac disease by Manfredi et al. [109]. Leitgeb et al. studied and investigated the immobilization of TGM onto surface-modified MNPs with carboxymethyl dextran for cleaner production technologies. In this case, the TGM was hyperactivated and exhibited 99% immobilization efficiency with 110% residual activity. It showed excellent thermal stability at 50 °C and at 70 °C [110]. Another TGM investigation by the same group reported on the synthesis of TGM immobilized in the form of CLEAs and magnetic CLEAs (mCLEAs). The TGM was precipitated in 2-propanol and later crosslinked with glutaraldehyde (GA), which resulted in 63% and 73% of residual activity for TGM CLEAs and mCLEAs, respectively. The CLEAs and mCLEAs showed great immobilization efficiency as well (95% and 90% for CLEAs and mCLEAs, respectively) [111]. Zhou et al. reported on TGM immobilized covalently on thermo-responsive carboxylated poly(N-isopropylacrylamide), where the immobilized TGM exhibited reversible solubility in an aqueous solution with a low critical solution temperature of 39 °C. Such immobilized TGM can be used to modify proteins in food processing and biomedical engineering [112]. The Wen-qiong report showed immobilization of TGM on an ultrafiltration polyethersulfone membrane surface, where it retained 50% of residual activity after 20 days. Additionally, the TGM-immobilized membrane had a higher relative membrane flux of 0.15 MPa in a membrane reactor [113].

4.2. Food Related Industries

As proteins are important food components, which play an important role in the phyisicochemical properties of food, the usage of TGM to crosslink food proteins to change their functional characteristics has been in progress for more than 30 years [114,115,116]. Enzymatic preparations of TGM have an important role in the food industry due to their practical utilization. Many reports describe the use of TGM in various food-related industries for the crosslinking of proteins, as in meat, cheese, yogurt, or bread (Figure 5).
It can also be used to produce composite edible films. TGM catalyzes the formation of crosslinks within a molecule as well as between molecules of other proteins. This feature impacts the changes in protein functionalities, such as solubility, foaming, emulsifying capacity, and gelation. As TGM has broad substrate specificity, Table 4 shows the reactivity of microbial TGM, which was investigated on different types of proteins that were derived from various foods [117,118,119,120].

4.2.1. Dairy Industry

TGM uses proteins such as casein or whey proteins as substrates to improve the foaming, emulsifying, and gelling properties of different foods. Casein is a major milk protein, which is a great substrate for TGM in dairy products due to its low degree of tertiary structure, flexibility, and the absence of disulfide bonds, which allows the exposure of reactive groups to TGM. On the other hand, the globular whey proteins, which do contain disulfide bonds, are poor substrates for TGM in the crosslinking process and therefore require modifications. Such modifications can be performed with reducing agents or increasing the pH value. They can also be achieved by heat denaturation or application of high hydrostatic pressure. However, such alterations in treatment can affect the interactions between the enzyme TGM in the TGM inhibitors that are present in milk serum and can also induce denaturation and result in cleavage of disulfide bonds, which later leads to unfolding of the proteins [26]. For example, the enzymatic crosslinking of casein was more resistant to digestion in comparison to the non-crosslinked casein. This suggests that the development of new types of products can offer carious food with improved structural characteristics, such as the polymerization of milk proteins with TGM results in protein film formation, which improves the functional properties of dairy products [121,122].
In the dairy industry, TGM has been introduced in many products. In yoghurts, it is used to prevent syneresis and for texture firming or softening since TGM-modified casein allows the manufacture of dairy products with a more consistent structure. The result of this reduced syneresis is a firmer and more homogeneous product. Various methods and protocols were also carried out investigating the use of TGM to increase cheese yield while enhancing the quality of low-fat cheese. TGM is also used in cheese manufacturing, where three methods are performed, including TGM:
-
the addition of TGM to milk, followed by heating for pasteurization and deactivation of enzymes, concluded with the addition of rennet to the milk;
-
the addition of rennet to the milk, followed by the addition of TGM;
-
the addition of TGM and rennet at the same time.
However, the reported investigations confirmed that the addition of TGM before the rennet prevented the coagulation of milk, while the simultaneous addition of both resulted in reduced resistance and hardness of the cheese [123]. By improving the cheese yield, textural properties, and its water-holding capacity, the use of TGM in cheese production is crucial. Other investigations reported on the improved heat stability and consistency of processed cheese after implementing TGM into the production [124,125,126,127,128].
Microbial TGM was used to treat the rheological and microstructural properties of yoghurt, where it was applied to milk before the fermentation. The TGM-mediated treatment decreased the ropiness of yoghurts and contributed to the acceptability of their texture, a study by Marhons suggests [129]. Salunke et al. investigated the use of micellar casein concentrate and milk proteins that were treated with TGM in different imitation cheese products. As TGM has the potential to modify the surface properties of milk protein concentrate and micellar casein concentrate, it may also improve functionality in imitation cheese, such as mozzarella [130]. Another study by the same group investigated the melt and stretch properties of dairy-based imitation mozzarella cheese, where the effect was studied of TGM-treated concentrates. The results demonstrated that TGM treatment modifies the investigated stretch and melt functionalities of milk protein concentrate and micellar casein concentrate [95]. Another study by Monsalve-Atencio et al. investigated the effect of TGM and its interaction with another enzyme, phospholipase, on the composition, yield, texture, and microstructure of semi-soft fresh cheese. The interaction of TGM with phospholipase showed the highest content of moisture in cheese value, which suggests an economically improved application of TGM in cheesemaking [131]. The addition of microbial TGM in quark cheese was studied, where the physicochemical, textural, sensory, and microbial properties of cheese were studied as well [132].

4.2.2. Baking Industry

The use of TGM in the baking industry is improving the quality of flour, and, consequently, the texture and volume of bread as proteins from grains are good substrates for crosslinkers by TGM. For example, rice flour is known to contain valuable nutrients, such as proteins, vitamins B and E, as well as fiber. However, it can only be used in and is limited to non-fermented bakery products. Investigations showed that the addition of TGM to rice flour improved the rheological properties of dough, and, by that, increased the content of triglycerides [133,134,135,136]. Similar studies were reported concerning cassava and wheat flour [137,138]. A TGM-induced protein aggregation method to improve the baking properties was investigated by Beck et al. [139], where the effect was studied of microbial TGM on the properties of rye dough. It was reported that the addition of TGM modified the rheological properties of rye flour dough, which resulted in a progressive increase in shear modulus. The increased TG concentration also showed an increase in crumb springiness and hardness, which demonstrated the improved breadmaking with the use of TGM. Another study also reports on improved rheological properties of gluten-free batter with the implementation of TGM. The crumb properties revealed that increased TGM concentration increased crumb chewiness and firmness [140]. The use of varying amounts of TGM also improved the baking quality of high-level sun pest wheat, where it was observed that TGM plays an important role in the baking quality. Increasing TGM activity caused increased bread characteristics of wheat, such as bread yield, height, crumb softness, pore structure, as well as decreased weight loss and wideness of the bread samples. The study concluded that the addition of TGM can restore the properties of bread and improve its overall protein structure [141]. Lang et al. evaluated the influence of TGM on the technological properties of gluten-free cakes of brown, black, and red rice. The effect of baking on the phenolic compound content was investigated as well [135].

4.2.3. Meat Industry

Numerous reports and studies are available investigating the use of TGM in meat products as one of the most widespread applications of TGM is in the restructuring of meat. Despite improving the structure and texture of the meat product, the use of TGM also provides cohesion without thermal processing or any additives, such as phosphates [59,142,143,144,145]. Studies show that the crosslinking activity of TGM in meat depends strongly on the temperature, pH, protein surface charge, and ionic strength. It was shown to improve other characteristics as well, such as water-binding, gelation, and emulsion stability. With the use of TGM in meat production, the secondary structure of a myosin heavy chain is changed by reduced α-helix content and increasing β-sheet content, which results in the formation of high-molecular-weight polymers. With such structural modifications, strong gels were formed with compact structural properties, which allow cohesiveness and improve the hardness of the meat. In addition, some studies also show that different degrees of gelation can be observed when induced by the addition of TGM [146,147,148]. This is also valid for fish skin gelatin. Namely, TGM-modified cold-water fish skin gelatin could be a potential mammalian gelatin replacer [149]. The addition of TGM also improves the quality of collagen, blood proteins, and provides higher nutritional value by supplementing with respective amino acids, such as exogenous lysine. TGM has allowed the production of new meat products, which use lower-quality raw materials instead of high-value meat products. The impact of TGM on the protein of such raw materials (skimmed milk powder or soy powder) does not alter the appearance, smell, texture, or nutritional value from products made with high-quality meat [150]. A study by Ribeiro et al. produced bovine meat with different levels of TGM combined with papain. The effects were investigated on pH, water activity, instrumental color, proximate composition, texture, and yield. It was concluded that the addition of TGM increased the yield of meat loafs [147]. In an interesting study by Wen et al., the enzyme TGM was used to develop 3D-printable meat analogs that imitate the physiochemical properties of beef. The TGM improved the rheological properties of raw meat, and provided a method for modifying the texture of meat analogs using TGM catalysis [151].
As the majority of the population are omnivores, TGM is being used widely in plant-based (PB) food industries, which are designing PB products that mimic the look, taste, and feel of animal-sourced foods. Zhou et al. developed PB protein gels for meat analogues that are created using slats, polysaccharides, and crosslinking enzymes, such as TGM, to modulate their gelation and assembly properties. In their study, the TGM increased the gel strength by forming covalent crosslinks between the potato protein molecules with more meat-like structures [152]. Additionally, traditional sausage production technologies can be used for PB analogues, such as a PB salami-type sausage analogue, which was manufactured with TGM-mediated soy protein isolate gels as binders, investigated in a study by Herz et al. [153].
In addition, the use of TGM is gaining much attention in the field of food packaging products; e.g., hemp proteins were used as raw material to obtain biodegradable films since they were demonstrated to act as both acyl donor and acceptor substrates of microbial TGM crosslinking [154,155]. Because such bioplastics show higher gas permeability and greater hydrophobicity, they may be useful as packaging systems for protecting food products from physical contamination and, thus, for extending their shelf-life. In the crosslinked gelatin-based films, food preservatives such as lysozyme or nisin may be incorporated to extend the shelf life of perishable foods. It was demonstrated that microbial-TGM-crosslinked gelatin-based films incorporated with lysozyme can control the release of this food preservative effectively.

4.3. Biotechnology and Cosmetics

Microbial TGM is an interesting tool for protein modification, which catalyzes protein crosslinking through isopeptide bond formation, which occurs between γ-carboxamide groups of glutamine residues, which include the acyl donor, and primary amines, such as ε-amino groups of lysine residues, which include the acyl acceptor. Therefore, research on TGM use can be applied to biomedical, biomaterial, cosmetic, and feedstock technologies. TGM can be used for modification of gelatin hydrogels and collagen for enhancing binding in different tissues. Microbial TGM was used to prepare collagen-grafted chitosan, which could serve not only to reduce the loss of moisture but also to absorb the moisture. With such properties, it showed the potentiality to repair skin in the cosmetic, biomedical, and pharmaceutical fields [156]. TGM-crosslinked whey proteins were also used to prepare a D-limonene emulsion, which can solve the problems of easy oxidation and poor water solubility of D-limonene. Limonene is an important ingredient in the formulation of different cosmetic and personal care products, such as aftershave lotions, bath products, cleansing products, eye shadows, hair products, lipsticks, shampoos, etc. [157]. Moreover, microbial TGM, as a protein-crosslinking enzyme in the processing of hair, improved the rigidity of hair fibers by 15.64% compared to a control when it was applied to damaged hair [158].
It has also been applied for bioconjugation, in order to create antibody–drug conjugates for various therapeutic applications. Additionally, new uses for TGM in the field of novel biomaterials are suggested and can be generated via site-specific substrate binding by proteins modified by TGM [159,160]. Regarding different feedstocks, TGM has shown to improve the physical properties of fish feed, while, in cosmetics, the TGM-catalyzed reactions between amino groups of starch and γ-carboxamide groups of collagen peptides were investigated and reported to increase the effectiveness of some synthesized materials, such as drugs [156,161,162].
Fusion proteins with dual functions are important for immunochemical assays. Among such assays are the enzyme-linked immunosorbent assay (ELISA) and Western blot assays. In that manner, genetic fusion provides poor yields when large-sized hybrid molecules are assembled. Therefore, TGM catalysis is considered as a method for the preparation of protein–protein conjugates. It was demonstrated that the coupling of two functional proteins, namely peroxidase and protein G, is possible through lysine and glutamine active sites. As a result, only a small amount of the desired conjugate was yielded. Later, when TGM-mediated conjugation was performed, only the desired conjugates were obtained [163,164]. This finding provided TGM the recognition that respective tags can be applied to recombinant production to terminal and internal sites. Therefore, acyl donor and acceptor incorporation enable covalent linkage in the monomeric subunits, which enhances the thermal stability of the dimer [165]. Native antibody site-specific modification enhances the properties of antibody-based bioconjugates. However, such antibodies have a single functionality. A work by Walker et al. addressed this limitation by designing heterofunctional substrates for microbial TGM that can contain both azide and methyltetrazine “click handles”, which present a powerful method in the toolbox for native antibody modification [166,167,168,169,170]. Antibody–drug conjugates for cancer treatment have placed site-specific TGM-catalyzed conjugation with cytotoxic properties at the very pinnacle of research. TGM’s remarkable properties make it a versatile tool for post-translational modification of various proteins (Figure 6). A few examples are PEGylation of small-protein drugs to elevate their half-life, or immobilization of biocatalysts that are prone to aggregation in order to increase their stability or covalent attachment of nucleic acids to proteins for combining the properties of both biomolecules [171,172,173,174]. The reactivity of microbial TGM was investigated at intrinsic lysine and glutamine sites of different antibodies [167,170,175,176,177,178]. The amino component in N-terminal pentaglycyl was found to mediate protein modification by TGM, which was performed by crosslinking of the enhanced green fluorescent protein. Many other protein–protein conjugates were performed using TGM-catalyzed conjugation. As reported by Bhokisham et al., attachment on solid supports occurred, which caused optimization of the reaction stoichiometry [179].
Covalent coupling of nucleic acid macromolecules, such as DNA and RNA, to proteins is a powerful method in molecular biology since both components exhibit excellent functions [180,181,182]. Therefore, protein–oligonucleotide conjugates became valuable tools in analytic and biomedical applications, for example, in drug delivery [183] and molecular diagnostics [184].
Protein–oligonucleotide ligation often relies on chemical methods that include functional groups of the desired target protein. As a result, partial functional loss may occur due to the steric hindrance or protein treatment with organic solvents. Therefore, site-specific coupling is performed to avoid residue modification, which is essential for protein function. In that manner, innovative strategies were developed to include microbial TGM in the modification process. Some research reports show aminated DNA, which was attached chemically to the acyl donor substrate and further coupled to alkaline phosphatase [185].
A modern technique to improve the pharmacokinetic properties of pharmacophore is PEGylation, where polyethylene glycol is attached to small-protein-based drugs. PEGylated pharmaceutics usually reduce immunogenicity compared to the non-PEGylated counterparts due to the enlarged hydrodynamic radius and higher conformational stability [186]. With increased bioavailability, the intravenous administration of such protein-based drugs can become more patient-friendly. The controlled conjugation strategies are very important since the modifications at binding interfaces or active sites of residues can affect the in vivo functions. Leading to undesired reactivity during conjugation, it can lead to a heterogeneous mixture of products with changed pharmacokinetic properties. Nevertheless, PEGylated proteins that are synthesized by chemical derivatization are already available on the market [186]. However, compared to chemical modification of such proteins, the site selectivity of microbial TGM is accessing PEGylated derivatives of various drugs without altering their properties [187].
Another way to manipulate specificity is covalent immobilization of TGM on solid supports. In a study by Grigoletto et al., TGM was coupled to agarose beads through an N-terminus to investigate its activity and substrate specificity. The immobilized TGM exhibited changed enzymatic activity and kinetic parameters, which were the result of chemical modification. Due to PEGylation, the immobilized TGM appeared more site-selective [171]. A biodegradable alternative to PEG, hydroxyethyl starch, is a polymeric molecule, which was used for microbial TGM-mediated protein conjugation and served as an acyl donor/acceptor to ligate monodansyl cadaverine [188]. Moreover, microbial TGM was also proved to be able to catalyze an acyltransfer reaction between the aminated oligosaccharides and acyl donor molecules [189], while glycosylation of catalase and trypsin was obtained by the transamidation of carboxamide functions.
Antibody–drug conjugates are promising tools for tumor treatments, where a chemotherapeutic is bound covalently to immunoglobulin, with the intent to enlarge the therapeutic range with the combination of powerful organic toxins and targeted specificity antibodies. Cytotoxic drugs are used widely to treat malignancies and solid tumors, and have, under specific clinical conditions, altered the natural course of certain diseases. Due to their intrinsic mode of active site, they are effective but can also cause significant on-target events that could result in the discontinuation of medication, which would increase the risk of recurrence of the tumor. For maintaining the efficiency of chemotherapeutics, efforts were undertaken to investigate novel approaches. Among such approaches was the conjugation of cytotoxic agents to antibodies. Many studies describe improved pharmacokinetics, enhanced efficacy, and reduced toxicity of antibody–drug conjugates when they are ligated site-selectively [190]. Non-directional methods generate heterogeneous products with statistically distributed coupling sites and fluctuating hydrophobic profiles, while site-specific conjugation provides reproducible hydrophobic properties with antibody modification restrictions (Figure 7).
Most of the antibody–drug conjugates in clinical use nowadays are assembled by random modification of cysteines and lysines. Many studies where an enzymatic approach was developed to modify antibodies site-selectively were reported [191,192,193,194,195,196]. Such an approach includes additional cysteine or selenocysteine residues [197,198]. Microbial TGM shows incapability of labelling glutamines in native human antibodies efficiently, although many Gln sites are exposed and available [199]. Therefore, a specific recognition tag is being incorporated and is reported to be a powerful strategy in overcoming such limitations. Research was performed at Pfizer, where a TGM recognition sequence was placed at various surface-exposed regions of a specific growth factor receptor antibody, which facilitated efficient labeling by microbial TGM [165].
In the synthesis process of an antibody–drug conjugate, all aspects must be taken into consideration. The target, antigen, antibody, linker, and the cytotoxic load must be evaluated for the targeted cancer indication. The antibodies with cytotoxic load must obtain excellent targeting capabilities to distinguish between healthy and tumor cells. In the process, antibodies that are engineered to follow a specific tumor antigen attach themselves to the surface of tumor cells. When processing within lysosomes or endosomes takes place, the antibody–drug conjugate releases its lethal load and destroys the targeted tumor cells (Figure 8). Due to its highly targeted tumor antigen expression, recognition, and requirements for effective internalization and processing, the antibody–drug conjugates are believed to provide a broader therapeutic opportunity in the ever-growing field of enzyme-mediated modification strategies [168].

5. Conclusions

TGMs remain one of the most complex families of enzymes that possess various structural and functional properties in mammalians, non-mammalian eucaryotes, and in bacteria. As highly efficient enzymes with unique features, they are used in the development and improvement of many versatile products. The extraordinary applicability of TGMs in the formulation of different dairy, meat, and feedstock products has led to an incredible enhancement in the food industries, where they have been proven to be efficient and applicable tools for the development of new products. Aside from the extensive use of TGM in food-related and manufacturing industries, multiple achievements have contributed to biotechnological research, where it is used as an antibody–drug conjugate, and such research has become a promising field for its refinement. Site-specific conjugation is a widely developed strategy to customize the properties of target proteins towards various applications in pharmaceutical and biomedical applications, by loading tumor-specific antibodies with small cytotoxic molecules, in order to create antibody–drug conjugates. In recent years, numerous studies have reported on the considerable impact that enzyme TGM has in either immobilized or free form. For example, while compared to traditional conjugation methodologies, TGM-mediated catalysis has many beneficial advantages. TGM is a powerful component in the biotechnological field, and an important tool in the bioconjugation process. Furthermore, TGM has the potential to be useful in many non-food-related fields as well, where it will continue to contribute to new innovative products, including using novel technologies for enzyme modifications by protein engineering and for cleaner productions. Reducing the costs of production is essential and is aiming and guiding their applications on a larger scale in various industrial sectors, such as in the design of different biotechnological products.

Author Contributions

Conceptualization, K.V. and M.L.; investigation, K.V.; resources, K.V., M.L. and Ž.K.; writing—original draft preparation, K.V.; writing—review and editing, K.V. and M.L.; visualization, K.V.; supervision, M.L.; funding acquisition, K.V., M.L. and Ž.K. All authors have read and agreed to the published version of the manuscript.

Funding

The authors acknowledge the financial support by the Slovenian Research Agency Program P2-0046 “Separation Processes and Product Design” and Research Projects: J2-3037 “Bionanotechnology as a tool for stabilization and applications of bioactive substances from natural sources”, Z2-4431 “Functional biocomposites for biomedical and sustainable applications”, L2-4430 “Production, isolation and formulation of health beneficial substances from Helichrysum italicum for applications in cosmetic industry”.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

The authors acknowledge the Slovenian Research Agency.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kant Bhatia, S.; Vivek, N.; Kumar, V.; Chandel, N.; Thakur, M.; Kumar, D.; Yang, Y.-H.; Pugazendhi, A.; Kumar, G. Molecular Biology Interventions for Activity Improvement and Production of Industrial Enzymes. Bioresour. Technol. 2021, 324, 124596. [Google Scholar] [CrossRef]
  2. Kari, J.; Schaller, K.; Molina, G.A.; Borch, K.; Westh, P. The Sabatier Principle as a Tool for Discovery and Engineering of Industrial Enzymes. Curr. Opin. Biotechnol. 2022, 78, 102843. [Google Scholar] [CrossRef]
  3. Tarafdar, A.; Sirohi, R.; Gaur, V.K.; Kumar, S.; Sharma, P.; Varjani, S.; Pandey, H.O.; Sindhu, R.; Madhavan, A.; Rajasekharan, R.; et al. Engineering Interventions in Enzyme Production: Lab to Industrial Scale. Bioresour. Technol. 2021, 326, 124771. [Google Scholar] [CrossRef] [PubMed]
  4. Trevisol, T.C.; Henriques, R.O.; Souza, A.J.A.; Furigo, A. An Overview of the Use of Proteolytic Enzymes as Exfoliating Agents. J. Cosmet. Dermatol. 2022, 21, 3300–3307. [Google Scholar] [CrossRef] [PubMed]
  5. Basso, A.; Serban, S. Industrial Applications of Immobilized Enzymes—A Review. Mol. Catal. 2019, 479, 110607. [Google Scholar] [CrossRef]
  6. Zhang, K.; Qu, G.; Liu, W.; Sun, Z. Structure-function relationships of industrial enzymes. Sheng Wu Gong Cheng Xue Bao 2019, 35, 1806–1818. [Google Scholar] [CrossRef] [PubMed]
  7. Prasad, S.; Roy, I. Converting Enzymes into Tools of Industrial Importance. Recent Pat. Biotechnol. 2018, 12, 33–56. [Google Scholar] [CrossRef] [PubMed]
  8. Madhavan, A.; Arun, K.B.; Binod, P.; Sirohi, R.; Tarafdar, A.; Reshmy, R.; Kumar Awasthi, M.; Sindhu, R. Design of Novel Enzyme Biocatalysts for Industrial Bioprocess: Harnessing the Power of Protein Engineering, High Throughput Screening and Synthetic Biology. Bioresour. Technol. 2021, 325, 124617. [Google Scholar] [CrossRef]
  9. Basso, A.; Serban, S. Overview of Immobilized Enzymes’ Applications in Pharmaceutical, Chemical, and Food Industry. Methods Mol. Biol. 2020, 2100, 27–63. [Google Scholar] [CrossRef] [PubMed]
  10. Kaushal, J.; Mehandia, S.; Singh, G.; Raina, A.; Arya, S.K. Catalase Enzyme: Application in Bioremediation and Food Industry. Biocatal. Agric. Biotechnol. 2018, 16, 192–199. [Google Scholar] [CrossRef]
  11. Macek, B.; Forchhammer, K.; Hardouin, J.; Weber-Ban, E.; Grangeasse, C.; Mijakovic, I. Protein Post-Translational Modifications in Bacteria. Nat. Rev. Microbiol. 2019, 17, 651–664. [Google Scholar] [CrossRef] [PubMed]
  12. Chung, C.H.; Yoo, H.M. Emerging Role of Protein Modification by UFM1 in Cancer. Biochem. Biophys. Res. Commun. 2022, 633, 61–63. [Google Scholar] [CrossRef]
  13. Keenan, E.K.; Zachman, D.K.; Hirschey, M.D. Discovering the Landscape of Protein Modifications. Mol. Cell 2021, 81, 1868–1878. [Google Scholar] [CrossRef] [PubMed]
  14. Pan, S.; Chen, R. Pathological Implication of Protein Post-Translational Modifications in Cancer. Mol. Asp. Med. 2022, 86, 101097. [Google Scholar] [CrossRef] [PubMed]
  15. Miwa, N. Innovation in the Food Industry Using Microbial Transglutaminase: Keys to Success and Future Prospects. Anal. Biochem. 2020, 597, 113638. [Google Scholar] [CrossRef]
  16. Moulton, K.R.; Sadiki, A.; Koleva, B.N.; Ombelets, L.J.; Tran, T.H.; Liu, S.; Wang, B.; Chen, H.; Micheloni, E.; Beuning, P.J.; et al. Site-Specific Reversible Protein and Peptide Modification: Transglutaminase-Catalyzed Glutamine Conjugation and Bioorthogonal Light-Mediated Removal. Bioconjug. Chem. 2019, 30, 1617–1621. [Google Scholar] [CrossRef]
  17. Stender, E.G.P.; Koutina, G.; Almdal, K.; Hassenkam, T.; Mackie, A.; Ipsen, R.; Svensson, B. Isoenergic Modification of Whey Protein Structure by Denaturation and Crosslinking Using Transglutaminase. Food Funct. 2018, 9, 797–805. [Google Scholar] [CrossRef]
  18. Deweid, L.; Hadjabdelhafid-Parisien, A.; Lafontaine, K.; Rochet, L.N.C.; Kolmar, H.; Pelletier, J.N. Glutamine-Walking: Creating Reactive Substrates for Transglutaminase-Mediated Protein Labeling. Methods Enzymol. 2020, 644, 121–148. [Google Scholar] [CrossRef]
  19. Akbari, M.; Razavi, S.H.; Kieliszek, M. Recent Advances in Microbial Transglutaminase Biosynthesis and Its Application in the Food Industry. Trends Food Sci. Technol. 2021, 110, 458–469. [Google Scholar] [CrossRef]
  20. Giosafatto, C.V.L.; Al-Asmar, A.; Mariniello, L. Transglutaminase Protein Substrates of Food Interest. In Enzymes in Food Technology: Improvements and Innovations; Kuddus, M., Ed.; Springer: Singapore, 2018; pp. 293–317. ISBN 9789811319334. [Google Scholar]
  21. Fatima, S.W.; Khare, S.K. Current Insight and Futuristic Vistas of Microbial Transglutaminase in Nutraceutical Industry. Microbiol. Res. 2018, 215, 7–14. [Google Scholar] [CrossRef]
  22. Wilhelmus, M.M.M.; Jongenelen, C.A.; Bol, J.G.J.M.; Drukarch, B. Interaction between Tissue Transglutaminase and Amyloid-Beta: Protein-Protein Binding versus Enzymatic Crosslinking. Anal. Biochem. 2020, 592, 113578. [Google Scholar] [CrossRef]
  23. Alaneed, R.; Naumann, M.; Pietzsch, M.; Kressler, J. Microbial Transglutaminase-Mediated Formation of Erythropoietin-Polyester Conjugates. J. Biotechnol. 2022, 346, 1–10. [Google Scholar] [CrossRef]
  24. Wu, T.; Huang, H.; Sheng, Y.; Shi, H.; Min, Y.; Liu, Y. Transglutaminase Mediated PEGylation of Nanobodies for Targeted Nano-Drug Delivery. J. Mater. Chem. B 2018, 6, 1011–1017. [Google Scholar] [CrossRef] [PubMed]
  25. Dell’Olmo, E.; Gaglione, R.; Arciello, A.; Piccoli, R.; Cafaro, V.; Di Maro, A.; Ragucci, S.; Porta, R.; Giosafatto, C.V.L. Transglutaminase-Mediated Crosslinking of a Host Defence Peptide Derived from Human Apolipoprotein B and Its Effect on the Peptide Antimicrobial Activity. Biochim. Biophys. Acta Gen. Subj. 2021, 1865, 129803. [Google Scholar] [CrossRef] [PubMed]
  26. Romeih, E.; Walker, G. Recent Advances on Microbial Transglutaminase and Dairy Application. Trends Food Sci. Technol. 2017, 62, 133–140. [Google Scholar] [CrossRef]
  27. Zhang, J.; Li, T.; Chen, Q.; Liu, H.; Kaplan, D.L.; Wang, Q. Application of Transglutaminase Modifications for Improving Protein Fibrous Structures from Different Sources by High-Moisture Extruding. Food Res. Int. 2023, 166, 112623. [Google Scholar] [CrossRef] [PubMed]
  28. Matsumura, Y.; Lee, D.-S.; Mori, T. Molecular Weight Distributions of α-Lactalbumin Polymers Formed by Mammalian and Microbial Transglutaminases. Food Hydrocoll. 2000, 14, 49–59. [Google Scholar] [CrossRef]
  29. Jaros, D.; Partschefeld, C.; Henle, T.; Rohm, H. Transglutaminase in Dairy Products: Chemistry, Physics, Applications. J. Texture Stud. 2006, 37, 113–155. [Google Scholar] [CrossRef]
  30. Yokoyama, K.; Nio, N.; Kikuchi, Y. Properties and Applications of Microbial Transglutaminase. Appl. Microbiol. Biotechnol. 2004, 64, 447–454. [Google Scholar] [CrossRef]
  31. Raak, N.; Abbate, R.A.; Lederer, A.; Rohm, H.; Jaros, D. Size Separation Techniques for the Characterisation of Cross-Linked Casein: A Review of Methods and Their Applications. Separations 2018, 5, 14. [Google Scholar] [CrossRef] [Green Version]
  32. Guyot, C.; Kulozik, U. Effect of Transglutaminase-Treated Milk Powders on the Properties of Skim Milk Yoghurt. Int. Dairy J. 2011, 21, 628–635. [Google Scholar] [CrossRef]
  33. Velazquez-Dominguez, A.; Hiolle, M.; Abdallah, M.; Delaplace, G.; Peixoto, P.P.S. Transglutaminase Cross-Linking on Dairy Proteins: Functionalities, Patents, and Commercial Uses. Int. Dairy J. 2023, 143, 105688. [Google Scholar] [CrossRef]
  34. RCSB Protein Data Bank. RCSB PDB: Homepage. Available online: https://www.rcsb.org/ (accessed on 12 March 2023).
  35. Adrio, J.L.; Demain, A.L. Microbial Enzymes: Tools for Biotechnological Processes. Biomolecules 2014, 4, 117–139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Fernandes, C.G.; Plácido, D.; Lousa, D.; Brito, J.A.; Isidro, A.; Soares, C.M.; Pohl, J.; Carrondo, M.A.; Archer, M.; Henriques, A.O. Structural and Functional Characterization of an Ancient Bacterial Transglutaminase Sheds Light on the Minimal Requirements for Protein Cross-Linking. Biochemistry 2015, 54, 5723–5734. [Google Scholar] [CrossRef] [PubMed]
  37. Duarte, L.; Matte, C.R.; Bizarro, C.V.; Ayub, M.A.Z. Transglutaminases: Part I—Origins, Sources, and Biotechnological Characteristics. World J. Microbiol. Biotechnol. 2020, 36, 15. [Google Scholar] [CrossRef] [PubMed]
  38. Luciano, F.B.; Arntfield, S. Use of Transglutaminases in Foods and Potential Utilization of Plants as a Transglutaminase Source—Review. Biotemas 2012, 25, 1–11. [Google Scholar] [CrossRef] [Green Version]
  39. Zhuang, R.; Khosla, C. Substrates, Inhibitors, and Probes of Mammalian Transglutaminase 2. Anal. Biochem. 2020, 591, 113560. [Google Scholar] [CrossRef]
  40. Jeitner, T.M.; Kelly, J.M. Stabilization of Guinea Pig Transglutaminase 2 Solutions. Anal. Biochem. 2022, 657, 114885. [Google Scholar] [CrossRef]
  41. Kieliszek, M.; Misiewicz, A. Microbial Transglutaminase and Its Application in the Food Industry. A Review. Folia Microbiol. 2014, 59, 241–250. [Google Scholar] [CrossRef] [Green Version]
  42. Taghi Gharibzahedi, S.M.; Koubaa, M.; Barba, F.J.; Greiner, R.; George, S.; Roohinejad, S. Recent Advances in the Application of Microbial Transglutaminase Crosslinking in Cheese and Ice Cream Products: A Review. Int. J. Biol. Macromol. 2018, 107, 2364–2374. [Google Scholar] [CrossRef]
  43. Sobieszczuk-Nowicka, E.; Krzesłowska, M.; Legocka, J. Transglutaminases and Their Substrates in Kinetin-Stimulated Etioplast-to-Chloroplast Transformation in Cucumber Cotyledons. Protoplasma 2008, 233, 187–194. [Google Scholar] [CrossRef] [PubMed]
  44. Campos, A.; Carvajal-Vallejos, P.K.; Villalobos, E.; Franco, C.F.; Almeida, A.M.; Coelho, A.V.; Torné, J.M.; Santos, M. Characterisation of Zea mays L. Plastidial Transglutaminase: Interactions with Thylakoid Membrane Proteins. Plant Biol. 2010, 12, 708–716. [Google Scholar] [CrossRef] [PubMed]
  45. Duarte, L.S.; Matte, C.R.; Dall Cortivo, P.R.; Nunes, J.E.S.; Barsé, L.Q.; Bizarro, C.V.; Ayub, M.A.Z. Expression of Bacillus amyloliquefaciens Transglutaminase in Recombinant E. coli under the Control of a Bicistronic Plasmid System in DO-Stat Fed-Batch Bioreactor Cultivations. Braz. J. Microbiol. 2021, 52, 1225–1233. [Google Scholar] [CrossRef]
  46. Wang, X.; Zhao, B.; Du, J.; Xu, Y.; Zhu, X.; Zhou, J.; Rao, S.; Du, G.; Chen, J.; Liu, S. Active Secretion of a Thermostable Transglutaminase Variant in Escherichia coli. Microb. Cell Fact. 2022, 21, 74. [Google Scholar] [CrossRef] [PubMed]
  47. Sato, R.; Minamihata, K.; Ariyoshi, R.; Taniguchi, H.; Kamiya, N. Recombinant Production of Active Microbial Transglutaminase in E. coli by Using Self-Cleavable Zymogen with Mutated Propeptide. Protein Expr. Purif. 2020, 176, 105730. [Google Scholar] [CrossRef] [PubMed]
  48. Kikuchi, Y.; Date, M.; Yokoyama, K.; Umezawa, Y.; Matsui, H. Secretion of Active-Form Streptoverticillium mobaraense Transglutaminase by Corynebacterium glutamicum: Processing of the pro-Transglutaminase by a Cosecreted Subtilisin-like Protease from Streptomyces albogriseolus. Appl. Environ. Microbiol. 2003, 69, 358–366. [Google Scholar] [CrossRef] [Green Version]
  49. Kashiwagi, T.; Yokoyama, K.-I.; Ishikawa, K.; Ono, K.; Ejima, D.; Matsui, H.; Suzuki, E. Crystal Structure of Microbial Transglutaminase from Streptoverticillium mobaraense. J. Biol. Chem. 2002, 277, 44252–44260. [Google Scholar] [CrossRef] [Green Version]
  50. Date, M.; Yokoyama, K.; Umezawa, Y.; Matsui, H.; Kikuchi, Y. Production of Native-Type Streptoverticillium mobaraense Transglutaminase in Corynebacterium glutamicum. Appl. Environ. Microbiol. 2003, 69, 3011–3014. [Google Scholar] [CrossRef] [Green Version]
  51. Pasternack, R.; Dorsch, S.; Otterbach, J.T.; Robenek, I.R.; Wolf, S.; Fuchsbauer, H.L. Bacterial Pro-Transglutaminase from Streptoverticillium mobaraense—Purification, Characterisation and Sequence of the Zymogen. Eur. J. Biochem. 1998, 257, 570–576. [Google Scholar] [CrossRef] [Green Version]
  52. Guerra-Rodríguez, E.; Vázquez, M. Evaluation of a Novel Low-Cost Culture Medium Containing Exclusively Milk, Potato and Glycerol for Microbial Transglutaminase Production by Streptomyces mobaraensis. Chem. Eng. Res. Des. 2014, 92, 784–791. [Google Scholar] [CrossRef]
  53. Hirono-Hara, Y.; Yui, M.; Hara, K.Y. Production of Transglutaminase in Glutathione-Producing Recombinant Saccharomyces Cerevisiae. AMB Express 2021, 11, 13. [Google Scholar] [CrossRef] [PubMed]
  54. Wang, H.; Zhang, Y.; Yuan, Z.; Zou, X.; Ji, Y.; Hou, J.; Zhang, J.; Lu, F.; Liu, Y. Crosslinking Mechanism on a Novel Bacillus Cereus Transglutaminase-Mediated Conjugation of Food Proteins. Foods 2022, 11, 3722. [Google Scholar] [CrossRef]
  55. Santhi, D.; Kalaikannan, A.; Malairaj, P.; Arun Prabhu, S. Application of Microbial Transglutaminase in Meat Foods: A Review. Crit. Rev. Food Sci. Nutr. 2017, 57, 2071–2076. [Google Scholar] [CrossRef]
  56. Ohtake, K.; Mukai, T.; Iraha, F.; Takahashi, M.; Haruna, K.-I.; Date, M.; Yokoyama, K.; Sakamoto, K. Engineering an Automaturing Transglutaminase with Enhanced Thermostability by Genetic Code Expansion with Two Codon Reassignments. ACS Synth. Biol. 2018, 7, 2170–2176. [Google Scholar] [CrossRef] [PubMed]
  57. Liu, Y.; Huang, L.; Zheng, D.; Fu, Y.; Shan, M.; Li, Y.; Xu, Z.; Jia, L.; Wang, W.; Lu, F. Characterization of Transglutaminase from Bacillus subtilis and Its Cross-Linking Function with a Bovine Serum Albumin Model. Food Funct. 2018, 9, 5560–5568. [Google Scholar] [CrossRef] [PubMed]
  58. Fu, L.; Wang, Y.; Ju, J.; Cheng, L.; Xu, Y.; Yu, B.; Wang, L. Extracellular Production of Active-Form Streptomyces mobaraensis Transglutaminase in Bacillus subtilis. Appl. Microbiol. Biotechnol. 2020, 104, 623–631. [Google Scholar] [CrossRef]
  59. Wang, S.; Yang, Z.; Li, Z.; Tian, Y. Heterologous Expression of Recombinant Transglutaminase in Bacillus subtilis SCK6 with Optimized Signal Peptide and Codon, and Its Impact on Gelatin Properties. J. Microbiol. Biotechnol. 2020, 30, 1082–1091. [Google Scholar] [CrossRef]
  60. Liu, Y.; Zhang, Y.; Guo, Z.; Wang, C.; Kang, H.; Li, J.; Wang, W.; Li, Y.; Lu, F.; Liu, Y. Enhancing the Functional Characteristics of Soy Protein Isolate via Cross-Linking Catalyzed by Bacillus subtilis Transglutaminase. J. Sci. Food Agric. 2021, 101, 4154–4160. [Google Scholar] [CrossRef]
  61. Salis, B.; Spinetti, G.; Scaramuzza, S.; Bossi, M.; Saccani Jotti, G.; Tonon, G.; Crobu, D.; Schrepfer, R. High-Level Expression of a Recombinant Active Microbial Transglutaminase in Escherichia coli. BMC Biotechnol. 2015, 15, 84. [Google Scholar] [CrossRef] [Green Version]
  62. Steffen, W.; Ko, F.C.; Patel, J.; Lyamichev, V.; Albert, T.J.; Benz, J.; Rudolph, M.G.; Bergmann, F.; Streidl, T.; Kratzsch, P.; et al. Discovery of a Microbial Transglutaminase Enabling Highly Site-Specific Labeling of Proteins. J. Biol. Chem. 2017, 292, 15622–15635. [Google Scholar] [CrossRef] [Green Version]
  63. Giordano, D.; Langini, C.; Caflisch, A.; Marabotti, A.; Facchiano, A. Molecular Dynamics Analysis of the Structural Properties of the Transglutaminases of Kutzneria albida and Streptomyces mobaraensis. Comput. Struct. Biotechnol. J. 2022, 20, 3924–3934. [Google Scholar] [CrossRef]
  64. Mottahedeh, J.; Marsh, R. Characterization of 101-KDa Transglutaminase from Physarum polycephalum and Identification of LAV1-2 as Substrate. J. Biol. Chem. 1998, 273, 29888–29895. [Google Scholar] [CrossRef] [Green Version]
  65. Klein, J.D.; Guzman, E.; Kuehn, G.D. Purification and Partial Characterization of Transglutaminase from Physarum polycephalum. J. Bacteriol. 1992, 174, 2599–2605. [Google Scholar] [CrossRef] [Green Version]
  66. Wada, F.; Nakamura, A.; Masutani, T.; Ikura, K.; Maki, M.; Hitomi, K. Identification of Mammalian-Type Transglutaminase in Physarum polycephalum. Evidence from the CDNA Sequence and Involvement of GTP in the Regulation of Transamidating Activity. Eur. J. Biochem. 2002, 269, 3451–3460. [Google Scholar] [CrossRef] [PubMed]
  67. Wada, F.; Hasegawa, H.; Nakamura, A.; Sugimura, Y.; Kawai, Y.; Sasaki, N.; Shibata, H.; Maki, M.; Hitomi, K. Identification of Substrates for Transglutaminase in Physarum polycephalum, an Acellular Slime Mold, upon Cellular Mechanical Damage. FEBS J. 2007, 274, 2766–2777. [Google Scholar] [CrossRef]
  68. Milani, A.; Vecchietti, D.; Rusmini, R.; Bertoni, G. TgpA, a Protein with a Eukaryotic-like Transglutaminase Domain, Plays a Critical Role in the Viability of Pseudomonas aeruginosa. PLoS ONE 2012, 7, e50323. [Google Scholar] [CrossRef] [Green Version]
  69. Ando, H.; Adachi, M.; Umeda, K.; Matsuura, A.; Nonaka, M.; Uchio, R.; Tanaka, H.; Motoki, M. Purification and Characteristics of a Novel Transglutaminase Derived from Microorganisms. Agric. Biol. Chem. 1989, 53, 2613–2617. [Google Scholar] [CrossRef] [Green Version]
  70. Fatima, S.W.; Khare, S.K. Effect of Key Regulators in Augmenting Transcriptional Expression of Transglutaminase in Streptomyces mobaraensis. Bioresour. Technol. 2021, 340, 125627. [Google Scholar] [CrossRef] [PubMed]
  71. Huang, Y.; Jin, M.; Yan, W.; Wu, Q.; Niu, Y.; Zou, C.; Jia, C.; Chang, Z.; Huang, J.; Jiang, D.; et al. A Point Mutant in the Promoter of Transglutaminase Gene Dramatically Increased Yield of Microbial Transglutaminase from Streptomyces mobaraensis TX1. Process Biochem. 2022, 112, 92–97. [Google Scholar] [CrossRef]
  72. Wang, H.; Chen, H.; Li, Q.; Yu, F.; Yan, Y.; Liu, S.; Tian, J.; Tan, J. Enhancing the Thermostability of Transglutaminase from Streptomyces mobaraensis Based on the Rational Design of a Disulfide Bond. Protein Expr. Purif. 2022, 195–196, 106079. [Google Scholar] [CrossRef] [PubMed]
  73. Cui, L.; Du, G.; Zhang, D.; Chen, J. Thermal Stability and Conformational Changes of Transglutaminase from a Newly Isolated Streptomyces hygroscopicus. Bioresour. Technol. 2008, 99, 3794–3800. [Google Scholar] [CrossRef] [PubMed]
  74. Zhang, D.; Wang, M.; Wu, J.; Cui, L.; Du, G.; Chen, J. Two Different Proteases from Streptomyces hygroscopicus Are Involved in Transglutaminase Activation. J. Agric. Food Chem. 2008, 56, 10261–10264. [Google Scholar] [CrossRef] [PubMed]
  75. Du, K.; Liu, Z.; Cui, W.; Zhou, L.; Liu, Y.; Du, G.; Chen, J.; Zhou, Z. PH-Dependent Activation of Streptomyces hygroscopicus Transglutaminase Mediated by Intein. Appl. Environ. Microbiol. 2014, 80, 723–729. [Google Scholar] [CrossRef] [PubMed]
  76. Liu, S.; Wan, D.; Wang, M.; Madzak, C.; Du, G.; Chen, J. Overproduction of Pro-Transglutaminase from Streptomyces hygroscopicus in Yarrowia lipolytica and Its Biochemical Characterization. BMC Biotechnol. 2015, 15, 75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Wan, W.; He, D.; Xue, Z.; Zhang, Z. Specific Mutation of Transglutaminase Gene from Streptomyces hygroscopicus H197 and Characterization of Microbial Transglutaminase. J. Biosci. 2017, 42, 537–546. [Google Scholar] [CrossRef] [PubMed]
  78. Chen, K.; Zhang, D.; Liu, S.; Wang, N.S.; Wang, M.; Du, G.; Chen, J. Improvement of Transglutaminase Production by Extending Differentiation Phase of Streptomyces hygroscopicus: Mechanism and Application. Appl. Microbiol. Biotechnol. 2013, 97, 7711–7719. [Google Scholar] [CrossRef]
  79. Ho, M.-L.; Leu, S.-Z.; Hsieh, J.-F.; Jiang, S.-T. Technical Approach to Simplify the Purification Method and Characterization of Microbial Transglutaminase Produced from Streptoverticillium ladakanum. J. Food Sci. 2000, 65, 76–80. [Google Scholar] [CrossRef]
  80. Lin, Y.-S.; Chao, M.-L.; Liu, C.-H.; Chu, W.-S. Cloning and Expression of the Transglutaminase Gene from Streptoverticillium ladakanum in Streptomyces lividans. Process Biochem. 2004, 39, 591–598. [Google Scholar] [CrossRef]
  81. Umezawa, Y.; Ohtsuka, T.; Yokoyama, K.; Nio, N. Comparison of Enzymatic Properties of Microbial Transglutaminase from Streptomyces sp. Food Sci. Technol. Res. 2002, 8, 113–118. [Google Scholar] [CrossRef] [Green Version]
  82. Langston, J.; Blinkovsky, A.; Byun, T.; Terribilini, M.; Ransbarger, D.; Xu, F. Substrate Specificity of Streptomyces Transglutaminases. Appl. Biochem. Biotechnol. 2007, 136, 291–308. [Google Scholar] [CrossRef]
  83. Sannasimuthu, A.; Ramani, M.; Paray, B.A.; Pasupuleti, M.; Al-Sadoon, M.K.; Alagumuthu, T.S.; Al-Mfarij, A.R.; Arshad, A.; Mala, K.; Arockiaraj, J. Arthrospira platensis Transglutaminase Derived Antioxidant Peptide-Packed Electrospun Chitosan/Poly (Vinyl Alcohol) Nanofibrous Mat Accelerates Wound Healing, in Vitro, via Inducing Mouse Embryonic Fibroblast Proliferation. Colloids Surf. B Biointerfaces 2020, 193, 111124. [Google Scholar] [CrossRef] [PubMed]
  84. Lin, S.-J.; Hsieh, Y.-F.; Lai, L.-A.; Chao, M.-L.; Chu, W.-S. Characterization and Large-Scale Production of Recombinant Streptoverticillium platensis Transglutaminase. J. Ind. Microbiol. Biotechnol. 2008, 35, 981–990. [Google Scholar] [CrossRef] [PubMed]
  85. Lerner, A.; Aminov, R.; Matthias, T. Transglutaminases in Dysbiosis as Potential Environmental Drivers of Autoimmunity. Front. Microbiol. 2017, 8, 66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Noda, S.; Miyazaki, T.; Tanaka, T.; Ogino, C.; Kondo, A. Production of Streptoverticillium cinnamoneum Transglutaminase and Cinnamic Acid by Recombinant Streptomyces lividans Cultured on Biomass-Derived Carbon Sources. Bioresour. Technol. 2012, 104, 648–651. [Google Scholar] [CrossRef]
  87. Duran, R.; Junqua, M.; Schmitter, J.M.; Gancet, C.; Goulas, P. Purification, Characterisation, and Gene Cloning of Transglutaminase from Streptoverticillium cinnamoneum CBS 683.68. Biochimie 1998, 80, 313–319. [Google Scholar] [CrossRef] [PubMed]
  88. Date, M.; Yokoyama, K.-I.; Umezawa, Y.; Matsui, H.; Kikuchi, Y. High Level Expression of Streptomyces mobaraensis Transglutaminase in Corynebacterium glutamicum Using a Chimeric Pro-Region from Streptomyces cinnamoneus Transglutaminase. J. Biotechnol. 2004, 110, 219–226. [Google Scholar] [CrossRef]
  89. Gharibzahedi, S.M.T.; Altintas, Z. Transglutaminase-Induced Free-Fat Yogurt Gels Supplemented with Tarragon Essential Oil-Loaded Nanoemulsions: Development, Optimization, Characterization, Bioactivity, and Storability. Gels 2022, 8, 551. [Google Scholar] [CrossRef]
  90. Akal, C.; Koçak, C.; Kanca, N.; Özer, B. Utilization of Reconstituted Whey Powder and Microbial Transglutaminase in Ayran (Drinking Yogurt) Production. Food Technol. Biotechnol. 2022, 60, 253–265. [Google Scholar] [CrossRef]
  91. Abou-Soliman, N.H.I.; Sakr, S.S.; Awad, S. Physico-Chemical, Microstructural and Rheological Properties of Camel-Milk Yogurt as Enhanced by Microbial Transglutaminase. J. Food Sci. Technol. 2017, 54, 1616–1627. [Google Scholar] [CrossRef] [Green Version]
  92. Li, H.; Cui, Y.; Zhang, L.; Zhang, L.; Liu, H.; Yu, J. Optimization of Recombinant Zea mays Transglutaminase Production and Its Influence on the Functional Properties of Yogurt. Food Sci. Biotechnol. 2017, 26, 723–730. [Google Scholar] [CrossRef]
  93. D’Alessandro, A.G.; Martemucci, G.; Faccia, M. Effects of Microbial Transglutaminase Levels on Donkey Cheese Production. J. Dairy Res. 2021, 88, 351–356. [Google Scholar] [CrossRef] [PubMed]
  94. Darnay, L.; Miklós, G.; Lőrincz, A.; Szakmár, K.; Pásztor-Huszár, K.; Laczay, P. Possible Inhibitory Effect of Microbial Transglutaminase on the Formation of Biogenic Amines during Trappist Cheese Ripening. Food Addit. Contam. Part A Chem. Anal. Control Expo. Risk Assess. 2022, 39, 580–587. [Google Scholar] [CrossRef] [PubMed]
  95. Salunke, P.; Marella, C.; Amamcharla, J.K.; Muthukumarappan, K.; Metzger, L.E. Use of Micellar Casein Concentrate and Milk Protein Concentrate Treated with Transglutaminase in Imitation Cheese Products-Melt and Stretch Properties. J. Dairy Sci. 2022, 105, 7904–7916. [Google Scholar] [CrossRef] [PubMed]
  96. D’Alessandro, A.G.; Martemucci, G.; Loizzo, P.; Faccia, M. Production of Cheese from Donkey Milk as Influenced by Addition of Transglutaminase. J. Dairy Sci. 2019, 102, 10867–10876. [Google Scholar] [CrossRef] [PubMed]
  97. Duarte, L.; Matte, C.R.; Bizarro, C.V.; Ayub, M.A.Z. Review Transglutaminases: Part II—Industrial Applications in Food, Biotechnology, Textiles and Leather Products. World J. Microbiol. Biotechnol. 2019, 36, 11. [Google Scholar] [CrossRef]
  98. Tesfaw, A.; Assefa, F. Applications of Transglutaminase in Textile, Wool, and Leather Processing. Int. J. Text. Sci. 2014, 3, 64–69. [Google Scholar]
  99. Arana-Peña, S.; Carballares, D.; Morellon-Sterlling, R.; Berenguer-Murcia, Á.; Alcántara, A.R.; Rodrigues, R.C.; Fernandez-Lafuente, R. Enzyme Co-Immobilization: Always the Biocatalyst Designers’ Choice…or Not? Biotechnol. Adv. 2021, 51, 107584. [Google Scholar] [CrossRef]
  100. Caparco, A.A.; Dautel, D.R.; Champion, J.A. Protein Mediated Enzyme Immobilization. Small 2022, 18, e2106425. [Google Scholar] [CrossRef]
  101. Federsel, H.-J.; Moody, T.S.; Taylor, S.J.C. Recent Trends in Enzyme Immobilization—Concepts for Expanding the Biocatalysis Toolbox. Molecules 2021, 26, 2822. [Google Scholar] [CrossRef]
  102. Rodrigues, R.C.; Berenguer-Murcia, Á.; Carballares, D.; Morellon-Sterling, R.; Fernandez-Lafuente, R. Stabilization of Enzymes via Immobilization: Multipoint Covalent Attachment and Other Stabilization Strategies. Biotechnol. Adv. 2021, 52, 107821. [Google Scholar] [CrossRef]
  103. Liu, Q.; Xun, G.; Feng, Y. The State-of-the-Art Strategies of Protein Engineering for Enzyme Stabilization. Biotechnol. Adv. 2019, 37, 530–537. [Google Scholar] [CrossRef]
  104. Chapman, R.; Stenzel, M.H. All Wrapped up: Stabilization of Enzymes within Single Enzyme Nanoparticles. J. Am. Chem. Soc. 2019, 141, 2754–2769. [Google Scholar] [CrossRef] [PubMed]
  105. Reyes-De-Corcuera, J.I.; Olstad, H.E.; García-Torres, R. Stability and Stabilization of Enzyme Biosensors: The Key to Successful Application and Commercialization. Annu. Rev. Food Sci. Technol. 2018, 9, 293–322. [Google Scholar] [CrossRef]
  106. Silva, C.; Martins, M.; Jing, S.; Fu, J.; Cavaco-Paulo, A. Practical Insights on Enzyme Stabilization. Crit. Rev. Biotechnol. 2018, 38, 335–350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Fatima, S.W.; Barua, S.; Sardar, M.; Khare, S.K. Immobilization of Transglutaminase on Multi-Walled Carbon Nanotubes and Its Application as Bioinspired Hydrogel Scaffolds. Int. J. Biol. Macromol. 2020, 163, 1747–1758. [Google Scholar] [CrossRef] [PubMed]
  108. Giannetto, M.; Mattarozzi, M.; Umiltà, E.; Manfredi, A.; Quaglia, S.; Careri, M. An Amperometric Immunosensor for Diagnosis of Celiac Disease Based on Covalent Immobilization of Open Conformation Tissue Transglutaminase for Determination of Anti-TTG Antibodies in Human Serum. Biosens. Bioelectron. 2014, 62, 325–330. [Google Scholar] [CrossRef] [PubMed]
  109. Manfredi, A.; Mattarozzi, M.; Giannetto, M.; Careri, M. Piezoelectric Immunosensor Based on Antibody Recognition of Immobilized Open-Tissue Transglutaminase: An Innovative Perspective on Diagnostic Devices for Celiac Disease. Sens. Actuators B Chem. 2014, 201, 300–307. [Google Scholar] [CrossRef]
  110. Gajšek, M.; Jančič, U.; Vasić, K.; Knez, Ž.; Leitgeb, M. Enhanced Activity of Immobilized Transglutaminase for Cleaner Production Technologies. J. Clean. Prod. 2019, 240, 118218. [Google Scholar] [CrossRef]
  111. Hojnik Podrepšek, G.; Knez, Ž.; Leitgeb, M. The Synthesis of (Magnetic) Crosslinked Enzyme Aggregates with Laccase, Cellulase, β-Galactosidase and Transglutaminase. Front. Bioeng. Biotechnol. 2022, 10, 813919. [Google Scholar] [CrossRef]
  112. Zhou, J.Q.; He, T.; Wang, J.W. The Microbial Transglutaminase Immobilization on Carboxylated Poly(N-Isopropylacrylamide) for Thermo-Responsivity. Enzym. Microb. Technol. 2016, 87–88, 44–51. [Google Scholar] [CrossRef]
  113. Wang, W.; Zhang, X. Optimization of Transglutaminase (TG) Immobilization on the Surface of Polyethersulfone Ultrafiltration Membrane and Its Characteristics in a Membrane Reactor. J. Biotechnol. 2018, 287, 41–51. [Google Scholar] [CrossRef]
  114. Ikura, K.; Kometani, T.; Yoshikawa, M.; Sasaki, R.; Chiba, H. Crosslinking of Casein Components by Transglutaminase. Agric. Biol. Chem. 1980, 44, 1567–1573. [Google Scholar] [CrossRef] [Green Version]
  115. Ikura, K.; Yoshikawa, M.; Sasaki, R.; Chiba, H. Incorporation of Amino Acids into Food Proteins by Transglutaminase. Agric. Biol. Chem. 1981, 45, 2587–2592. [Google Scholar] [CrossRef] [Green Version]
  116. Ikura, K.; Kometani, T.; Sasaki, R.; Chiba, H. Crosslinking of Soybean 7S and 11S Proteins by Transglutaminase. Agric. Biol. Chem. 1980, 44, 2979–2984. [Google Scholar] [CrossRef]
  117. Zhang, Y.; Simpson, B.K. Food-Related Transglutaminase Obtained from Fish/Shellfish. Crit. Rev. Food Sci. Nutr. 2020, 60, 3214–3232. [Google Scholar] [CrossRef]
  118. Matthias, T.; Jeremias, P.; Neidhöfer, S.; Lerner, A. The Industrial Food Additive, Microbial Transglutaminase, Mimics Tissue Transglutaminase and Is Immunogenic in Celiac Disease Patients. Autoimmun. Rev. 2016, 15, 1111–1119. [Google Scholar] [CrossRef] [PubMed]
  119. Lerner, A.; Matthias, T. Possible Association between Celiac Disease and Bacterial Transglutaminase in Food Processing: A Hypothesis. Nutr. Rev. 2015, 73, 544–552. [Google Scholar] [CrossRef]
  120. Aaron, L.; Torsten, M. Microbial Transglutaminase: A New Potential Player in Celiac Disease. Clin. Immunol. 2019, 199, 37–43. [Google Scholar] [CrossRef] [PubMed]
  121. Gauche, C.; Barreto, P.L.M.; Bordignon-Luiz, M.T. Effect of Thermal Treatment on Whey Protein Polymerization by Transglutaminase: Implications for Functionality in Processed Dairy Foods. LWT—Food Sci. Technol. 2010, 43, 214–219. [Google Scholar] [CrossRef]
  122. Jaros, D.; Schwarzenbolz, U.; Raak, N.; Löbner, J.; Henle, T.; Rohm, H. Corrigendum to “Cross-Linking with Microbial Transglutaminase: Relationship between Polymerisation Degree and Stiffness of Acid Casein Gels” [Int Dairy J 38 (2014) 174–178]. Int. Dairy J. 2014, 39, 345–347. [Google Scholar] [CrossRef]
  123. Mahmood, W.A. Effect of Microbial Transglutaminase Treatment on Soft Cheese Properties. Mesop. J. Agric. 2009, 37, 19–27. [Google Scholar] [CrossRef] [Green Version]
  124. Aaltonen, T.; Huumonen, I.; Myllärinen, P. Controlled Transglutaminase Treatment in Edam Cheese-Making. Int. Dairy J. 2014, 38, 179–182. [Google Scholar] [CrossRef]
  125. Özer, B.; Hayaloglu, A.A.; Yaman, H.; Gürsoy, A.; Şener, L. Simultaneous Use of Transglutaminase and Rennet in White-Brined Cheese Production. Int. Dairy J. 2013, 33, 129–134. [Google Scholar] [CrossRef]
  126. Fotschki, J.; Wróblewska, B.; Fotschki, B.; Kalicki, B.; Rigby, N.; Mackie, A. Microbial Transglutaminase Alters the Immunogenic Potential and Cross-Reactivity of Horse and Cow Milk Proteins. J. Dairy Sci. 2020, 103, 2153–2166. [Google Scholar] [CrossRef]
  127. Hovjecki, M.; Miloradovic, Z.; Mirkovic, N.; Radulovic, A.; Pudja, P.; Miocinovic, J. Rheological and Textural Properties of Goat’s Milk Set-Type Yoghurt as Affected by Heat Treatment, Transglutaminase Addition and Storage. J. Sci. Food Agric. 2021, 101, 5898–5906. [Google Scholar] [CrossRef] [PubMed]
  128. Han, Y.; Mei, Y.; Li, K.; Xu, Y.; Wang, F. Effect of Transglutaminase on Rennet-Induced Gelation of Skim Milk and Soymilk Mixtures. J. Sci. Food Agric. 2019, 99, 1820–1827. [Google Scholar] [CrossRef]
  129. Marhons, Š.; Hyršlová, I.; Stetsenko, V.; Jablonská, E.; Veselý, M.; Míchová, H.; Čurda, L.; Štĕtina, J. Properties of Yoghurt Treated with Microbial Transglutaminase and Exopolysaccharides. Int. Dairy J. 2023, 144, 105701. [Google Scholar] [CrossRef]
  130. Salunke, P.; Marella, C.; Amamcharla, J.K.; Muthukumarappan, K.; Metzger, L.E. Use of Micellar Casein Concentrate and Milk Protein Concentrate Treated with Transglutaminase in Imitation Cheese Products—Unmelted Texture. J. Dairy Sci. 2022, 105, 7891–7903. [Google Scholar] [CrossRef]
  131. Monsalve-Atencio, R.; Sanchez-Soto, K.; Chica, J.; Camaño Echavarría, J.A.; Vega-Castro, O. Interaction between Phospholipase and Transglutaminase in the Production of Semi-Soft Fresh Cheese and Its Effect on the Yield, Composition, Microstructure and Textural Properties. LWT 2022, 154, 112722. [Google Scholar] [CrossRef]
  132. Seyed-Moslemi, S.A.; Hesari, J.; Peighambardoust, S.H.; Peighambardoust, S.J. Effect of Microbial Lipase and Transglutaminase on the Textural, Physicochemical, and Microbial Parameters of Fresh Quark Cheese. J. Dairy Sci. 2021, 104, 7489–7499. [Google Scholar] [CrossRef]
  133. Grossmann, I.; Döring, C.; Jekle, M.; Becker, T.; Koehler, P. Compositional Changes and Baking Performance of Rye Dough As Affected by Microbial Transglutaminase and Xylanase. J. Agric. Food Chem. 2016, 64, 5751–5758. [Google Scholar] [CrossRef] [PubMed]
  134. Mazzeo, M.F.; Bonavita, R.; Maurano, F.; Bergamo, P.; Siciliano, R.A.; Rossi, M. Biochemical Modifications of Gliadins Induced by Microbial Transglutaminase on Wheat Flour. Biochim. Biophys. Acta 2013, 1830, 5166–5174. [Google Scholar] [CrossRef] [PubMed]
  135. Lang, G.H.; Kringel, D.H.; Acunha, T.D.S.; Ferreira, C.D.; Dias, Á.R.G.; Zavareze, E.D.R.; de Oliveira, M. Cake of Brown, Black and Red Rice: Influence of Transglutaminase on Technological Properties, in Vitro Starch Digestibility and Phenolic Compounds. Food Chem. 2020, 318, 126480. [Google Scholar] [CrossRef]
  136. Altındağ, G.; Certel, M.; Erem, F.; İlknur Konak, Ü. Quality Characteristics of Gluten-Free Cookies Made of Buckwheat, Corn, and Rice Flour with/without Transglutaminase. Food Sci. Technol. Int. 2015, 21, 213–220. [Google Scholar] [CrossRef]
  137. Gharibzahedi, S.M.T.; Yousefi, S.; Chronakis, I.S. Microbial Transglutaminase in Noodle and Pasta Processing. Crit. Rev. Food Sci. Nutr. 2019, 59, 313–327. [Google Scholar] [CrossRef]
  138. Kiyat, W.E.; Christopher, A.; Rianti, A.; Pari, R.F. Application of Transglutaminase in Developing Cassava-Based Wet Noodle for Quality and Shelf Life Improvement: A Review. Recent Pat. Food Nutr. Agric. 2020, 11, 229–234. [Google Scholar] [CrossRef]
  139. Beck, M.; Jekle, M.; Selmair, P.L.; Koehler, P.; Becker, T. Rheological Properties and Baking Performance of Rye Dough as Affected by Transglutaminase. J. Cereal Sci. 2011, 54, 29–36. [Google Scholar] [CrossRef]
  140. Onyango, C.; Mutungi, C.; Unbehend, G.; Lindhauer, M.G. Rheological and Baking Characteristics of Batter and Bread Prepared from Pregelatinised Cassava Starch and Sorghum and Modified Using Microbial Transglutaminase. J. Food Eng. 2010, 97, 465–470. [Google Scholar] [CrossRef]
  141. Dizlek, H.; Özer, M.S. Improvement Bread Characteristics of High Level Sunn Pest (Eurygaster integriceps) Damaged Wheat by Using Transglutaminase and Some Additives. J. Cereal Sci. 2017, 77, 90–96. [Google Scholar] [CrossRef]
  142. Kim, H.; Lee, M.-Y.; Lee, J.; Jo, Y.-J.; Choi, M.-J. Effects of Glucono-δ-Lactone and Transglutaminase on the Physicochemical and Textural Properties of Plant-Based Meat Patty. Foods 2022, 11, 3337. [Google Scholar] [CrossRef] [PubMed]
  143. Kaić, A.; Janječić, Z.; Žgur, S.; Šikić, M.; Potočnik, K. Physicochemical and Sensory Attributes of Intact and Restructured Chicken Breast Meat Supplemented with Transglutaminase. Animals 2021, 11, 2641. [Google Scholar] [CrossRef]
  144. Kaufmann, A.; Köppel, R.; Widmer, M. Determination of Microbial Transglutaminase in Meat and Meat Products. Food Addit. Contam. Part A Chem. Anal. Control Expo. Risk Assess. 2012, 29, 1364–1373. [Google Scholar] [CrossRef] [PubMed]
  145. Zinina, O.; Merenkova, S.; Galimov, D.; Okuskhanova, E.; Rebezov, M.; Khayrullin, M.; Anichkina, O. Effects of Microbial Transglutaminase on Technological, Rheological, and Microstructural Indicators of Minced Meat with the Addition of Plant Raw Materials. Int. J. Food Sci. 2020, 2020, 8869401. [Google Scholar] [CrossRef] [PubMed]
  146. Yang, X.; Zhang, Y. Expression of Recombinant Transglutaminase Gene in Pichia Pastoris and Its Uses in Restructured Meat Products. Food Chem. 2019, 291, 245–252. [Google Scholar] [CrossRef]
  147. Ribeiro, W.O.; Ozaki, M.M.; dos Santos, M.; de Castro, R.J.S.; Sato, H.H.; Câmara, A.K.F.I.; Rodríguez, A.P.; Campagnol, P.C.B.; Pollonio, M.A.R. Evaluating Different Levels of Papain as Texture Modifying Agent in Bovine Meat Loaf Containing Transglutaminase. Meat Sci. 2023, 198, 109112. [Google Scholar] [CrossRef] [PubMed]
  148. Tokay, F.G.; Alp, A.C.; Yerlikaya, P. Production and Shelf Life of Restructured Fish Meat Binded by Microbial Transglutaminase. LWT 2021, 152, 112369. [Google Scholar] [CrossRef]
  149. Xu, J.; Yang, L.; Nie, Y.; Yang, M.; Wu, W.; Wang, Z.; Wang, X.; Zhong, J. Effect of Transglutaminase Crosslinking on the Structural, Physicochemical, Functional, and Emulsion Stabilization Properties of Three Types of Gelatins. LWT 2022, 163, 113543. [Google Scholar] [CrossRef]
  150. Baugreet, S.; Kerry, J.P.; Brodkorb, A.; Gomez, C.; Auty, M.; Allen, P.; Hamill, R.M. Optimisation of Plant Protein and Transglutaminase Content in Novel Beef Restructured Steaks for Older Adults by Central Composite Design. Meat Sci. 2018, 142, 65–77. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Wen, Y.; Kim, H.W.; Park, H.J. Effects of Transglutaminase and Cooking Method on the Physicochemical Characteristics of 3D-Printable Meat Analogs. Innov. Food Sci. Emerg. Technol. 2022, 81, 103114. [Google Scholar] [CrossRef]
  152. Zhou, H.; Hu, X.; Xiang, X.; McClements, D.J. Modification of Textural Attributes of Potato Protein Gels Using Salts, Polysaccharides, and Transglutaminase: Development of Plant-Based Foods. Food Hydrocoll. 2023, 144, 108909. [Google Scholar] [CrossRef]
  153. Herz, E.; Kinne, T.; Terjung, N.; Gibis, M.; Weiss, J. Influence of Extrudate to SPI-Gel-Binder Ratios and Transglutaminase Crosslinking on Texture of a Plant-Based Salami Analogue. Future Foods 2023, 7, 100235. [Google Scholar] [CrossRef]
  154. Mirpoor, S.F.; Giosafatto, C.V.L.; Di Girolamo, R.; Famiglietti, M.; Porta, R. Hemp (Cannabis sativa) Seed Oilcake as a Promising by-Product for Developing Protein-Based Films: Effect of Transglutaminase-Induced Crosslinking. Food Packag. Shelf Life 2022, 31, 100779. [Google Scholar] [CrossRef]
  155. Sabaghi, M.; Joly, C.; Adt, I.; Ozturk, K.; Cottaz, A.; Degraeve, P. Effect of Crosslinking by Microbial Transglutaminase of Gelatin Films on Lysozyme Kinetics of Release in Food Simulants. Food Biosci. 2022, 48, 101816. [Google Scholar] [CrossRef]
  156. Fan, L.; Wu, H.; Zhou, X.; Peng, M.; Tong, J.; Xie, W.; Liu, S. Transglutaminase-Catalyzed Grafting Collagen on Chitosan and Its Characterization. Carbohydr. Polym. 2014, 105, 253–259. [Google Scholar] [CrossRef]
  157. Bilawal, A.; Gantumur, M.-A.; Huang, Y.; Qayum, A.; Ishfaq, M.; Jiang, Z. Effect of Transglutaminase Cross-Linking and Emulsification on the Stability and Digestion of Limonene Emulsions. Process Biochem. 2023, 131, 210–216. [Google Scholar] [CrossRef]
  158. Kim, Y.S.; Park, S.J. Application of Transglutaminase for Hair Revitalization. J. Soc. Cosmet. Sci. Korea 2013, 39, 25–30. [Google Scholar] [CrossRef]
  159. Wakabayashi, R.; Yahiro, K.; Hayashi, K.; Goto, M.; Kamiya, N. Protein-Grafted Polymers Prepared Through a Site-Specific Conjugation by Microbial Transglutaminase for an Immunosorbent Assay. Biomacromolecules 2017, 18, 422–430. [Google Scholar] [CrossRef]
  160. Collighan, R.J.; Griffin, M. Transglutaminase 2 Cross-Linking of Matrix Proteins: Biological Significance and Medical Applications. Amino Acids 2009, 36, 659–670. [Google Scholar] [CrossRef]
  161. Wen, H.; Hu, J.; Ge, H.; Zou, S.; Xiao, Y.; Li, Y.; Feng, H.; Fan, L. Preparation and Characterization of Aminoethyl Hydroxypropyl Starch Modified with Collagen Peptide. Int. J. Biol. Macromol. 2017, 101, 996–1003. [Google Scholar] [CrossRef]
  162. Moore Rosset, E.; Trombetta-eSilva, J.; Hepfer, G.; Chen, P.; Yao, H.; Bradshaw, A.D. Inhibition of Transglutaminase Activity in Periodontitis Rescues Periodontal Ligament Collagen Content and Architecture. J. Periodontal. Res. 2020, 55, 107–115. [Google Scholar] [CrossRef]
  163. Bechtold, U.; Otterbach, J.T.; Pasternack, R.; Fuchsbauer, H.L. Enzymic Preparation of Protein G-Peroxidase Conjugates Catalysed by Transglutaminase. J. Biochem. 2000, 127, 239–245. [Google Scholar] [CrossRef]
  164. Spolaore, B.; Damiano, N.; Raboni, S.; Fontana, A. Site-Specific Derivatization of Avidin Using Microbial Transglutaminase. Bioconjug. Chem. 2014, 25, 470–480. [Google Scholar] [CrossRef]
  165. Deweid, L.; Avrutina, O.; Kolmar, H. Microbial Transglutaminase for Biotechnological and Biomedical Engineering. Biol. Chem. 2019, 400, 257–274. [Google Scholar] [CrossRef] [PubMed]
  166. Walker, J.A.; Bohn, J.J.; Ledesma, F.; Sorkin, M.R.; Kabaria, S.R.; Thornlow, D.N.; Alabi, C.A. Substrate Design Enables Heterobifunctional, Dual “Click” Antibody Modification via Microbial Transglutaminase. Bioconjug. Chem. 2019, 30, 2452–2457. [Google Scholar] [CrossRef] [PubMed]
  167. Huggins, I.J.; Medina, C.A.; Springer, A.D.; van den Berg, A.; Jadhav, S.; Cui, X.; Dowdy, S.F. Site Selective Antibody-Oligonucleotide Conjugation via Microbial Transglutaminase. Molecules 2019, 24, 3287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Dickgiesser, S.; Deweid, L.; Kellner, R.; Kolmar, H.; Rasche, N. Site-Specific Antibody-Drug Conjugation Using Microbial Transglutaminase. Methods Mol. Biol. 2019, 2012, 135–149. [Google Scholar] [CrossRef]
  169. Ebenig, A.; Juettner, N.E.; Deweid, L.; Avrutina, O.; Fuchsbauer, H.-L.; Kolmar, H. Efficient Site-Specific Antibody-Drug Conjugation by Engineering a Nature-Derived Recognition Tag for Microbial Transglutaminase. Chembiochem 2019, 20, 2411–2419. [Google Scholar] [CrossRef]
  170. Schneider, H.; Deweid, L.; Avrutina, O.; Kolmar, H. Recent Progress in Transglutaminase-Mediated Assembly of Antibody-Drug Conjugates. Anal. Biochem. 2020, 595, 113615. [Google Scholar] [CrossRef]
  171. Grigoletto, A.; Mero, A.; Yoshioka, H.; Schiavon, O.; Pasut, G. Covalent Immobilisation of Transglutaminase: Stability and Applications in Protein PEGylation. J. Drug Target. 2017, 25, 856–864. [Google Scholar] [CrossRef]
  172. Zhou, J.Q.; He, T.; Wang, J.W. PEGylation of Cytochrome c at the Level of Lysine Residues Mediated by a Microbial Transglutaminase. Biotechnol. Lett. 2016, 38, 1121–1129. [Google Scholar] [CrossRef]
  173. Grigoletto, A.; Mero, A.; Maso, K.; Pasut, G. Transgultaminase-Mediated Nanoarmoring of Enzymes by PEGylation. Methods Enzymol. 2017, 590, 317–346. [Google Scholar] [CrossRef]
  174. Mero, A.; Spolaore, B.; Veronese, F.M.; Fontana, A. Transglutaminase-Mediated PEGylation of Proteins: Direct Identification of the Sites of Protein Modification by Mass Spectrometry Using a Novel Monodisperse PEG. Bioconjug. Chem. 2009, 20, 384–389. [Google Scholar] [CrossRef] [PubMed]
  175. Kaunisto, H.; Salmi, T.; Lindfors, K.; Kemppainen, E. Antibody Responses to Transglutaminase 3 in Dermatitis Herpetiformis: Lessons from Celiac Disease. Int. J. Mol. Sci. 2022, 23, 2910. [Google Scholar] [CrossRef] [PubMed]
  176. Anami, Y.; Tsuchikama, K. Transglutaminase-Mediated Conjugations. Methods Mol. Biol. 2020, 2078, 71–82. [Google Scholar] [CrossRef] [PubMed]
  177. Sato, K.; Nanri, K. Gluten Ataxia: Anti-Transglutaminase-6 Antibody as a New Biomarker. Brain Nerve 2017, 69, 933–940. [Google Scholar] [CrossRef]
  178. Ghosh, K.; Ghosh, K.; Agarwal, R.; Shah, K.; Mishra, K. Anti Tissue Transglutaminase Antibody in Idiopathic Autoimmune Haemolytic Anemia. Transfus. Apher. Sci. 2019, 58, 693–696. [Google Scholar] [CrossRef]
  179. Bhokisham, N.; Pakhchanian, H.; Quan, D.; Tschirhart, T.; Tsao, C.-Y.; Payne, G.F.; Bentley, W.E. Modular Construction of Multi-Subunit Protein Complexes Using Engineered Tags and Microbial Transglutaminase. Metab. Eng. 2016, 38, 1–9. [Google Scholar] [CrossRef]
  180. Takahara, M.; Wakabayashi, R.; Minamihata, K.; Goto, M.; Kamiya, N. Primary Amine-Clustered DNA Aptamer for DNA-Protein Conjugation Catalyzed by Microbial Transglutaminase. Bioconjug. Chem. 2017, 28, 2954–2961. [Google Scholar] [CrossRef]
  181. Kitaoka, M.; Tsuruda, Y.; Tanaka, Y.; Goto, M.; Mitsumori, M.; Hayashi, K.; Hiraishi, Y.; Miyawaki, K.; Noji, S.; Kamiya, N. Transglutaminase-Mediated Synthesis of a DNA-(Enzyme)n Probe for Highly Sensitive DNA Detection. Chemistry 2011, 17, 5387–5392. [Google Scholar] [CrossRef]
  182. Maki, K.; Shibata, T.; Kawabata, S.-I. Transglutaminase-Catalyzed Incorporation of Polyamines Masks the DNA-Binding Region of the Transcription Factor Relish. J. Biol. Chem. 2017, 292, 6369–6380. [Google Scholar] [CrossRef] [Green Version]
  183. Mascini, M.; Palchetti, I.; Tombelli, S. Nucleic Acid and Peptide Aptamers: Fundamentals and Bioanalytical Aspects. Angew. Chem. Int. Ed. Engl. 2012, 51, 1316–1332. [Google Scholar] [CrossRef] [PubMed]
  184. Zhao, D.; Kong, Y.; Zhao, S.; Xing, H. Engineering Functional DNA-Protein Conjugates for Biosensing, Biomedical, and Nanoassembly Applications. Top. Curr. Chem. 2020, 378, 41. [Google Scholar] [CrossRef] [PubMed]
  185. Tominaga, J.; Kemori, Y.; Tanaka, Y.; Maruyama, T.; Kamiya, N.; Goto, M. An Enzymatic Method for Site-Specific Labeling of Recombinant Proteins with Oligonucleotides. Chem. Commun. 2007, 4, 401–403. [Google Scholar] [CrossRef] [PubMed]
  186. Dozier, J.K.; Distefano, M.D. Site-Specific PEGylation of Therapeutic Proteins. Int. J. Mol. Sci. 2015, 16, 25831–25864. [Google Scholar] [CrossRef] [Green Version]
  187. Fontana, A.; Spolaore, B.; Mero, A.; Veronese, F.M. Site-Specific Modification and PEGylation of Pharmaceutical Proteins Mediated by Transglutaminase. Adv. Drug Deliv. Rev. 2008, 60, 13–28. [Google Scholar] [CrossRef]
  188. Besheer, A.; Hertel, T.C.; Kressler, J.; Mäder, K.; Pietzsch, M. Enzymatically Catalyzed HES Conjugation Using Microbial Transglutaminase: Proof of Feasibility. J. Pharm. Sci. 2009, 98, 4420–4428. [Google Scholar] [CrossRef]
  189. Villalonga, M.L.; Villalonga, R.; Mariniello, L.; Gómez, L.; Pierro, P.D.; Porta, R. Transglutaminase-Catalysed Glycosidation of Trypsin with Aminated Polysaccharides. World J. Microbiol. Biotechnol. 2006, 22, 595–602. [Google Scholar] [CrossRef] [Green Version]
  190. Bryant, P.; Pabst, M.; Badescu, G.; Bird, M.; McDowell, W.; Jamieson, E.; Swierkosz, J.; Jurlewicz, K.; Tommasi, R.; Henseleit, K.; et al. In Vitro and In Vivo Evaluation of Cysteine Rebridged Trastuzumab–MMAE Antibody Drug Conjugates with Defined Drug-to-Antibody Ratios. Mol. Pharm. 2015, 12, 1872–1879. [Google Scholar] [CrossRef]
  191. Walsh, S.J.; Bargh, J.D.; Dannheim, F.M.; Hanby, A.R.; Seki, H.; Counsell, A.J.; Ou, X.; Fowler, E.; Ashman, N.; Takada, Y.; et al. Site-Selective Modification Strategies in Antibody-Drug Conjugates. Chem. Soc. Rev. 2021, 50, 1305–1353. [Google Scholar] [CrossRef]
  192. Li, C.; Chong, G.; Zong, G.; Knorr, D.A.; Bournazos, S.; Aytenfisu, A.H.; Henry, G.K.; Ravetch, J.V.; MacKerell, A.D.; Wang, L.-X. Site-Selective Chemoenzymatic Modification on the Core Fucose of an Antibody Enhances Its Fcγ Receptor Affinity and ADCC Activity. J. Am. Chem. Soc. 2021, 143, 7828–7838. [Google Scholar] [CrossRef]
  193. Zhang, C.; Dai, P.; Vinogradov, A.A.; Gates, Z.P.; Pentelute, B.L. Site-Selective Cysteine-Cyclooctyne Conjugation. Angew. Chem. Int. Ed. Engl. 2018, 57, 6459–6463. [Google Scholar] [CrossRef] [Green Version]
  194. Haque, M.; Forte, N.; Baker, J.R. Site-Selective Lysine Conjugation Methods and Applications towards Antibody-Drug Conjugates. Chem. Commun. 2021, 57, 10689–10702. [Google Scholar] [CrossRef] [PubMed]
  195. Chowdhury, A.; Chatterjee, S.; Pongen, A.; Sarania, D.; Tripathi, N.M.; Bandyopadhyay, A. Site-Selective, Chemical Modification of Protein at Aromatic Side Chain and Their Emergent Applications. Protein Pept. Lett. 2021, 28, 788–808. [Google Scholar] [CrossRef] [PubMed]
  196. Marculescu, C.; Lakshminarayanan, A.; Gault, J.; Knight, J.C.; Folkes, L.K.; Spink, T.; Robinson, C.V.; Vallis, K.; Davis, B.G.; Cornelissen, B. Probing the Limits of Q-Tag Bioconjugation of Antibodies. Chem. Commun. 2019, 55, 11342–11345. [Google Scholar] [CrossRef] [Green Version]
  197. Drake, P.M.; Rabuka, D. Recent Developments in ADC Technology: Preclinical Studies Signal Future Clinical Trends. BioDrugs 2017, 31, 521–531. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  198. Li, X.; Nelson, C.G.; Nair, R.R.; Hazlehurst, L.; Moroni, T.; Martinez-Acedo, P.; Nanna, A.R.; Hymel, D.; Burke, T.R.; Rader, C. Stable and Potent Selenomab-Drug Conjugates. Cell Chem. Biol. 2017, 24, 433–442.e6. [Google Scholar] [CrossRef] [Green Version]
  199. Mindt, T.L.; Jungi, V.; Wyss, S.; Friedli, A.; Pla, G.; Novak-Hofer, I.; Grünberg, J.; Schibli, R. Modification of Different IgG1 Antibodies via Glutamine and Lysine Using Bacterial and Human Tissue Transglutaminase. Bioconjug. Chem. 2008, 19, 271–278. [Google Scholar] [CrossRef]
Figure 1. TGM-mediated reactions: (a) acyl transfer reaction, (b) protein crosslinking reaction, (c) deamidation.
Figure 1. TGM-mediated reactions: (a) acyl transfer reaction, (b) protein crosslinking reaction, (c) deamidation.
Ijms 24 12402 g001
Figure 2. TGM-promoted crosslinks based on low and high substrate concentrations that can produce proteins with new and unique functional properties.
Figure 2. TGM-promoted crosslinks based on low and high substrate concentrations that can produce proteins with new and unique functional properties.
Ijms 24 12402 g002
Figure 4. Production of TGM by microorganisms.
Figure 4. Production of TGM by microorganisms.
Ijms 24 12402 g004
Figure 5. Opportunities for applications of TGM in the food industry.
Figure 5. Opportunities for applications of TGM in the food industry.
Ijms 24 12402 g005
Figure 6. Biotechnological applications of microbial TGM—an overview.
Figure 6. Biotechnological applications of microbial TGM—an overview.
Ijms 24 12402 g006
Figure 7. Schematic variations of non-directed modification and site-specific modification of antibody–drug conjugates.
Figure 7. Schematic variations of non-directed modification and site-specific modification of antibody–drug conjugates.
Ijms 24 12402 g007
Figure 8. Schematic biological activity mechanism of antibody–drug conjugates.
Figure 8. Schematic biological activity mechanism of antibody–drug conjugates.
Ijms 24 12402 g008
Table 1. Industrial enzymes and their significant applications.
Table 1. Industrial enzymes and their significant applications.
EnzymeSubstrateIndustrial Application
AmylaseCarbohydrateDetergents, Paper and pulp, Textile, Baking, Starch, Fuel
LaccaseBenzenediolTextiles, Paper and pulp, Food
LipaseFat, oilDetergents, Oil and fat, Food and baking, Paper and pulp, Fine chemicals
Pectinase Pectin Food, Beverages, Textiles
Protease Protein, polypeptideDetergents, Food and Leather processing, Water treatment, Animal feeds
PullulanasePolysaccharideFood, Starch
TGMProtein, amineCosmetics, Textiles, Food
XylaseXylanAnimal feeds, Baking and food, Paper and pulp
Table 2. Properties and differences in TGMs from different sources.
Table 2. Properties and differences in TGMs from different sources.
ConditionTGM from Microbial SourcesTGM from Animal Sources
Temperature (°C)45–5550–55
pH5–86
Isoelectric point94.5
MW (kDa)37,80076,600
Table 3. Various microbial strain sources for the isolation of TGM.
Table 3. Various microbial strain sources for the isolation of TGM.
MicroorganismReference
Bacillus subtilis[57,58,59,60]
Escherichia coli[46,47,61]
Kutzneria albida[62,63]
Physarum polycephalum[64,65,66,67]
Pseudomonas aeruginosa[68]
Sterptoverticilliu mobaraensis[69,70,71,72]
Streptomyces hygroscopicus[73,74,75,76,77,78]
Streptomyces ladakanum[79,80]
Streptomyces libani[81]
Streptomyces nigrescens[82]
Streptomyces platensis[80,83,84]
Streptomyces sioyaensis[85]
Streptoverticillium cinnamoneum[86,87,88]
Table 4. Reactivity of microbial TGM in relation to different food proteins.
Table 4. Reactivity of microbial TGM in relation to different food proteins.
Food ProteinImproved Functional PropertiesReactivity
EggOvalbumin (egg white)Depending on condition
Egg yolk proteinWell
MeatMyoglobinDepending on condition
GelatinVery well
CollagenWell
MyosinVery well
ActinDoes not react
MilkCaseinVery well
α-lactalbuminDepending on condition
β-lactoglobulinDepending on condition
Sodium caseinateVery well
Soybean11S GlobulinVery well
7S globulinVery well
WheatGliadinWell
GluteninWell
Some examples of the TGMs used in the food industry are listed below.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Vasić, K.; Knez, Ž.; Leitgeb, M. Transglutaminase in Foods and Biotechnology. Int. J. Mol. Sci. 2023, 24, 12402. https://doi.org/10.3390/ijms241512402

AMA Style

Vasić K, Knez Ž, Leitgeb M. Transglutaminase in Foods and Biotechnology. International Journal of Molecular Sciences. 2023; 24(15):12402. https://doi.org/10.3390/ijms241512402

Chicago/Turabian Style

Vasić, Katja, Željko Knez, and Maja Leitgeb. 2023. "Transglutaminase in Foods and Biotechnology" International Journal of Molecular Sciences 24, no. 15: 12402. https://doi.org/10.3390/ijms241512402

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop